1
|
Nguyen K, Hsiao CHC, Jin Y, Wiemer AJ, Vinogradova O. Investigation of structural and dynamic properties of the Butyrophilin BTN3A1/BTN2A1 cytoplasmic complex by 19F solution NMR. FASEB J 2025; 39:e70449. [PMID: 40079188 PMCID: PMC11908674 DOI: 10.1096/fj.202402975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 02/24/2025] [Indexed: 03/14/2025]
Abstract
Butyrophilin 3A1 (BTN3A1) is an integral membrane protein capable of detecting phosphoantigens, like (E)-4-hydroxy-3-methyl-but-2-enyl diphosphate (HMBPP), through its internal B30.2 domain. Detection of phosphoantigens leads to interactions with butyrophilin 2A1 and the subsequent activation of γδ-T cells. Though crystallography and functional assays have been crucial for determining vital residues of the BTN3A1/HMBPP/BTN2A1 complex, the mechanism for signal transduction is still unclear. Here, we utilize 19F solution NMR to observe potential conformational and dynamic changes of specific residues upon complex formation. With point mutants of BTN3A1, we show that W421C, T449C, and T506C are residues that are influenced by HMBPP and BTN2A1 association, while T304C, G323C, C387, and C511 are not impacted. 19F labeling of W421C reduces the binding affinity of BTN2A1 toward BTN3A1/HMBPP, which indicates that W421 is located at the binding interface. T506 is located away from the phosphoantigen binding site, so its observable chemical shift perturbation suggests that there is a larger conformational change of the BTN3A1 B30.2 domain upon binding HMBPP and BTN2A1. The juxtamembrane residues, T304C, and G323C are not affected, showing that the changes are localized within the B30.2 domain of BTN3A1. Using BTN3A1 T449C, we were able to detect differential binding modes of synthetic HMBPP analogs, showing that it is possible to assess differences in protein conformations that are induced by binding of different ligands. Taken together, these findings illustrate the dynamic processes involved in phosphoantigen detection by the HMBPP receptor.
Collapse
Affiliation(s)
- Khiem Nguyen
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut, USA
| | | | - Yiming Jin
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut, USA
| | - Andrew J Wiemer
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut, USA
- Institute for Systems Genomics, University of Connecticut, Storrs, Connecticut, USA
| | - Olga Vinogradova
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut, USA
| |
Collapse
|
2
|
Saberi M, Dekkers R, Passerini L, Huber M, Overhand M, Ubbink M. Terminal spin labeling of xylotriose strongly affects interactions in the active site of xylanase BcX. JOURNAL OF BIOMOLECULAR NMR 2025:10.1007/s10858-025-00459-w. [PMID: 40072774 DOI: 10.1007/s10858-025-00459-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Paramagnetic probes provide long-range distance information and report on minor conformations of biomacromolecules. However, it is important to realize that any probe can affect the system of interest. Here, we report on the effects of attaching a small nitroxide spin label [TEMPO, (2,2,6,6-tetramethylpiperidin-1-yl)oxyl] to xylotriose, a substrate of the enzyme xylanase from Bacillus circulans (BcX). BcX has a long and narrow active site cleft accommodating six xylose units and a secondary binding site on its surface. The aim of the study was to probe the interactions of the substrate with the enzyme using paramagnetic relaxation enhancements (PREs). Binding of the substrate to the surface exposed secondary binding site resulted in strong and localized PREs, indicative of well-defined binding. The xylotriose with diamagnetic control tag was still able to bind the active site cleft, though the rate of exchange was reduced relative to that of untagged xylotriose. The substrate with the paramagnetic TEMPO was not able to bind inside the active site cleft. Also, additional interactions on another surface location showed differences between the paramagnetic substrate and the diamagnetic control, despite the minimal chemical differences between TEMPO modified xylotriose and its reduced, diamagnetic counterpart. Our findings underscore the sensitivity of BcX substrate binding to minor substrate modifications. This study serves as a reminder that any probe, including the attachment of a small paramagnetic group, can affect the behavior of the system under investigation. Even the chemical difference between a paramagnetic tag and its diamagnetic control can result in differences in the molecular interactions.
Collapse
Affiliation(s)
- Mahin Saberi
- Department of Macromolecular Biochemistry, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - René Dekkers
- Department of Macromolecular Biochemistry, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
- Department of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Leonardo Passerini
- Department of Physics, Huygens-Kamerlingh Onnes Laboratory, Leiden University, Niels Bohrweg 2, 2333 CA, Leiden, The Netherlands
| | - Martina Huber
- Department of Physics, Huygens-Kamerlingh Onnes Laboratory, Leiden University, Niels Bohrweg 2, 2333 CA, Leiden, The Netherlands
| | - Mark Overhand
- Department of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Marcellus Ubbink
- Department of Macromolecular Biochemistry, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.
| |
Collapse
|
3
|
Wang D, Wang J, Lang Y, Huang M, Hu S, Liu H, Sun B, Long Y, Wu J, Dong W. Interactions between food matrices and odorants: A review. Food Chem 2025; 466:142086. [PMID: 39612859 DOI: 10.1016/j.foodchem.2024.142086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/20/2024] [Accepted: 11/14/2024] [Indexed: 12/01/2024]
Abstract
Currently, although odorants of various foods have been thoroughly studied, the regulation of food aromas is still difficult due to the interaction between odorants and food matrices. These complex matrices in food may interact with odorants to change the volatility of odorants, which in turn affect food aroma. Clarifying the interaction between them are promising for predicting food aroma formation, which will provide valuable support for a high-efficiency food industry. Herein, the research progresses on interactions between food matrices and odorants are reviewed. First, the analysis methods and their advantages and disadvantages are introduced and discussed emphatically, including sensory-analysis methods, characterization methods of the volatility changes of odorants, and the research methods of interaction mechanism. Further, the research advances of interactions among proteins, carbohydrates, lipids, and polyphenols with odorants are summarized briefly. Finally, the existing problems are discussed and the research prospects are proposed.
Collapse
Affiliation(s)
- Danqing Wang
- Key Laboratory of Geriatric Nutrition and Health, (Beijing Technology and Business University), Ministry of Education, Beijing 100048, PR China; Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University (BTBU), Beijing, 100048, PR China
| | - Juan Wang
- Key Laboratory of Geriatric Nutrition and Health, (Beijing Technology and Business University), Ministry of Education, Beijing 100048, PR China; Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University (BTBU), Beijing, 100048, PR China
| | - Ying Lang
- Guizhou Wangmao Jiuqu Research Institute Co., Ltd., Guiyang, Guizhou 550081, PR China
| | - Mingquan Huang
- Key Laboratory of Geriatric Nutrition and Health, (Beijing Technology and Business University), Ministry of Education, Beijing 100048, PR China; Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University (BTBU), Beijing, 100048, PR China.
| | - Shenglan Hu
- Guizhou Wangmao Jiuqu Research Institute Co., Ltd., Guiyang, Guizhou 550081, PR China
| | - Hongqin Liu
- Key Laboratory of Geriatric Nutrition and Health, (Beijing Technology and Business University), Ministry of Education, Beijing 100048, PR China; Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University (BTBU), Beijing, 100048, PR China
| | - Baoguo Sun
- Key Laboratory of Geriatric Nutrition and Health, (Beijing Technology and Business University), Ministry of Education, Beijing 100048, PR China; Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University (BTBU), Beijing, 100048, PR China.
| | - Yao Long
- Guizhou Wangmao Jiuqu Research Institute Co., Ltd., Guiyang, Guizhou 550081, PR China
| | - Jihong Wu
- Key Laboratory of Geriatric Nutrition and Health, (Beijing Technology and Business University), Ministry of Education, Beijing 100048, PR China; Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University (BTBU), Beijing, 100048, PR China
| | - Wei Dong
- Key Laboratory of Geriatric Nutrition and Health, (Beijing Technology and Business University), Ministry of Education, Beijing 100048, PR China; Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University (BTBU), Beijing, 100048, PR China
| |
Collapse
|
4
|
Kratzwald S, Schwarz TC, Ledolter K, Hlavac M, Felkl M, Becker CFW, Konrat R, Lichtenecker RJ. Synthesis of a 13C/ 2H Labeled Building Block to Probe the Phosphotyrosine Interactome Using Biomolecular NMR Spectroscopy. Chembiochem 2025; 26:e202400663. [PMID: 39271462 PMCID: PMC11727006 DOI: 10.1002/cbic.202400663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/13/2024] [Accepted: 09/13/2024] [Indexed: 09/15/2024]
Abstract
Phosphotyrosine (pTyr) recognition coordinates the assembly of protein complexes, thus controlling key events of cell cycle, cell development and programmed cell death. Although many aspects of membrane receptor function and intracellular signal transduction have been deciphered in the last decades, the details of how phosphorylation alters protein-protein interaction and creates regulating switches of protein activity and localization often remains unclear. We developed a synthetic route to a protected phophotyrosine building block with isolated 13C-1H spins in the aromatic ring. The compound can be used for solid phase peptide synthesis (SPPS) and readily applied to study affinity, dynamics and interactions on an atomic level using NMR spectroscopy. As a first example, we prepared an isotopologue of a pTyr containing 12mer peptide (pY1021) as part of the platelet-derived growth factor to analyze the binding to the phospholipase C-γ (PLCγ-1) SH2 domain.
Collapse
Affiliation(s)
- Sarah Kratzwald
- Institute of Organic ChemistryFaculty of ChemistryUniversity of ViennaWähringerstr. 38Vienna1090Austria
- Mag-Lab ViennaKarl-Farkas Gasse 22Vienna1030Austria
| | - Thomas C. Schwarz
- Department of Structural and Computational BiologyUniversity of ViennaVienna Biocenter 5Vienna1030Austria
- Max Perutz LabsVienna Biocenter Campus (VBC)Vienna Biocenter 5Vienna1030Austria
- Christian Doppler Laboratory for High-Content Structural Biology and BiotechnologyDepartment of Structural and Computational BiologyMax Perutz LabsUniversity of ViennaVienna Biocenter 5Vienna1030Austria
| | - Karin Ledolter
- Department of Structural and Computational BiologyUniversity of ViennaVienna Biocenter 5Vienna1030Austria
- Max Perutz LabsVienna Biocenter Campus (VBC)Vienna Biocenter 5Vienna1030Austria
| | - Matus Hlavac
- Mag-Lab ViennaKarl-Farkas Gasse 22Vienna1030Austria
| | - Manuel Felkl
- Institute of Biological ChemistryFaculty of ChemistryUniversity of ViennaWähringerstr. 38Vienna1090Austria
| | - Christian F. W. Becker
- Institute of Biological ChemistryFaculty of ChemistryUniversity of ViennaWähringerstr. 38Vienna1090Austria
| | - Robert Konrat
- Department of Structural and Computational BiologyUniversity of ViennaVienna Biocenter 5Vienna1030Austria
- Max Perutz LabsVienna Biocenter Campus (VBC)Vienna Biocenter 5Vienna1030Austria
- Christian Doppler Laboratory for High-Content Structural Biology and BiotechnologyDepartment of Structural and Computational BiologyMax Perutz LabsUniversity of ViennaVienna Biocenter 5Vienna1030Austria
| | - Roman J. Lichtenecker
- Institute of Organic ChemistryFaculty of ChemistryUniversity of ViennaWähringerstr. 38Vienna1090Austria
- Mag-Lab ViennaKarl-Farkas Gasse 22Vienna1030Austria
- Christian Doppler Laboratory for High-Content Structural Biology and BiotechnologyDepartment of Structural and Computational BiologyMax Perutz LabsUniversity of ViennaVienna Biocenter 5Vienna1030Austria
| |
Collapse
|
5
|
Kirkman T, Dos Santos Silva C, Tosin M, Bertacine Dias MV. How to Find a Fragment: Methods for Screening and Validation in Fragment-Based Drug Discovery. ChemMedChem 2024; 19:e202400342. [PMID: 39198213 DOI: 10.1002/cmdc.202400342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 08/20/2024] [Accepted: 08/28/2024] [Indexed: 09/01/2024]
Abstract
Fragment-based drug discovery (FBDD) is a crucial strategy for developing new drugs that have been applied to diverse targets, from neglected infectious diseases to cancer. With at least seven drugs already launched to the market, this approach has gained interest in both academics and industry in the last 20 years. FBDD relies on screening small libraries with about 1000-2000 compounds of low molecular weight (about 300 Da) using several biophysical methods. Because of the reduced size of the compounds, the chemical space and diversity can be better explored than large libraries used in high throughput screenings. This review summarises the most common biophysical techniques used in fragment screening and orthogonal validation. We also explore the advantages and drawbacks of the different biophysical techniques and examples of applications and strategies.
Collapse
Affiliation(s)
- Tim Kirkman
- Department of Chemistry, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Catharina Dos Santos Silva
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, Av. Prof. Lineu Prestes, 1374, CEP 05508-000, São Paulo, SP, Brazil
| | - Manuela Tosin
- Department of Chemistry, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Marcio Vinicius Bertacine Dias
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, Av. Prof. Lineu Prestes, 1374, CEP 05508-000, São Paulo, SP, Brazil
| |
Collapse
|
6
|
Toscano G, Rosati M, Barbieri L, Maier K, Banci L, Luchinat E, Konrat R, Lichtenecker RJ. The synthesis of specifically isotope labelled fluorotryptophan and its use in mammalian cell-based protein expression for 19F-NMR applications. Chem Commun (Camb) 2024; 60:14188-14191. [PMID: 39512115 DOI: 10.1039/d4cc04789c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
19F nuclei serve as versatile sensors for detecting protein interactions and dynamics in biomolecular NMR spectroscopy. Although various methods have been developed to incorporate fluorine-containing aromatic residues into proteins using E. coli or cell-free expression techniques, similar approaches for protein production in mammalian cell lines remain limited. Here, we present a cost-effective synthetic route to obtain selectively deuterated, carbon-13 labeled fluorotryptophan and demonstrate its use in introducing 19F-13C spin pairs into carbonic anhydrase 2 and superoxide dismutase, following an expression protocol utilizing HEK cells.
Collapse
Affiliation(s)
- Giorgia Toscano
- Institute of Organic Chemistry, University of Vienna, Währinger Str. 38, 1090-Vienna, Austria.
- Christian Doppler Laboratory for High-Content Structural Biology and Biotechnology, Max Perutz Laboratories, University of Vienna, Campus Vienna Biocenter 5, 1030 Vienna, Austria
- Vienna Doctoral School in Chemistry (DoSChem), University of Vienna, Währingerstraße 42, 1090 Vienna, Austria
| | - Martina Rosati
- CERM Magnetic Resonance Center, Università degli Studi di Firenze, Sesto Fiorentino, Italy
| | - Letizia Barbieri
- CERM Magnetic Resonance Center, Università degli Studi di Firenze, Sesto Fiorentino, Italy
- Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine CIRMMP, Sesto Fiorentino, Italy
| | - Katharina Maier
- Institute of Organic Chemistry, University of Vienna, Währinger Str. 38, 1090-Vienna, Austria.
| | - Lucia Banci
- CERM Magnetic Resonance Center, Università degli Studi di Firenze, Sesto Fiorentino, Italy
- Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine CIRMMP, Sesto Fiorentino, Italy
- Dipartimento di Chimica, Università degli Studi di Firenze, Sesto Fiorentino, Italy
| | - Enrico Luchinat
- CERM Magnetic Resonance Center, Università degli Studi di Firenze, Sesto Fiorentino, Italy
- Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine CIRMMP, Sesto Fiorentino, Italy
- Dipartimento di Chimica, Università degli Studi di Firenze, Sesto Fiorentino, Italy
| | - Robert Konrat
- Christian Doppler Laboratory for High-Content Structural Biology and Biotechnology, Max Perutz Laboratories, University of Vienna, Campus Vienna Biocenter 5, 1030 Vienna, Austria
- Department of Structural and Computational Biology, Max Perutz Laboratories, University of Vienna, Dr-Bohr-Gasse 9, 1030-Vienna, Austria
- Mag-Lab, Karl-Farkas-Gasse 22, 1030 Vienna, Austria
| | - Roman J Lichtenecker
- Institute of Organic Chemistry, University of Vienna, Währinger Str. 38, 1090-Vienna, Austria.
- Christian Doppler Laboratory for High-Content Structural Biology and Biotechnology, Max Perutz Laboratories, University of Vienna, Campus Vienna Biocenter 5, 1030 Vienna, Austria
- Mag-Lab, Karl-Farkas-Gasse 22, 1030 Vienna, Austria
| |
Collapse
|
7
|
Vural O, Jololian L, Pan L. DeepLigType: Predicting Ligand Types of ProteinLigand Binding Sites Using a Deep Learning Model. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2024; PP:116-123. [PMID: 39509302 DOI: 10.1109/tcbb.2024.3493820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
The analysis of protein-ligand binding sites plays a crucial role in the initial stages of drug discovery. Accurately predicting the ligand types that are likely to bind to protein-ligand binding sites enables more informed decision making in drug design. Our study, DeepLigType, determines protein-ligand binding sites using Fpocket and then predicts the ligand type of these pockets with the deep learning model, Convolutional Block Attention Module (CBAM) with ResNet. CBAM-ResNet has been trained to accurately predict five distinct ligand types. We classified protein-ligand binding sites into five different categories according to the type of response ligands cause when they bind to their target proteins, which are antagonist, agonist, activator, inhibitor, and others. We created a novel dataset, referred to as LigType5, from the widely recognized PDBbind and scPDB dataset for training and testing our model. While the literature mostly focuses on the specificity and characteristic analysis of protein binding sites by experimental (laboratory-based) methods, we propose a computational method with the DeepLigType architecture. DeepLigType demonstrated an accuracy of 74.30% and an AUC of 0.83 in ligand type prediction on a novel test dataset using the CBAM-ResNet deep learning model.
Collapse
|
8
|
Bui DT, Kitova EN, Kitov PI, Han L, Mahal LK, Klassen JS. Deciphering Pathways and Thermodynamics of Protein Assembly Using Native Mass Spectrometry. J Am Chem Soc 2024; 146:28809-28821. [PMID: 39387708 DOI: 10.1021/jacs.4c08455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Protein oligomerization regulates many critical physiological processes, and its dysregulation can contribute to dysfunction and diseases. Elucidating the assembly pathways and quantifying their underlying thermodynamic and kinetic parameters are crucial for a comprehensive understanding of biological processes and for advancing therapeutics targeting abnormal protein oligomerization. Established binding assays, with limited mass precision, often rely on simplified models for data interpretation. In contrast, high-resolution native mass spectrometry (nMS) can directly determine the stoichiometry of biomolecular complexes in vitro. However, quantification is hindered by the fact that the relative abundances of gas-phase ions generally do not reflect solution concentrations due to nonuniform response factors. Recently, slow mixing mode (SLOMO)-nMS, which can quantify the relative response factors of interacting species, has been demonstrated to reliably measure the affinity (Kd) of binary biomolecular complexes. Here, we introduce an extended form of SLOMO-nMS that enables simultaneous quantification of the thermodynamics in multistep association reactions. Application of this method to homo-oligomerization of concanavalin A and insulin confirmed the reliability of the assay and uncovered details about the assembly processes that had previously resisted elucidation. Results acquired using SLOMO-nMS implemented with charge detection shed new light on the binding of recombinant human angiotensin-converting enzyme 2 and the SARS-CoV-2 spike protein. Importantly, new assembly pathways were uncovered, and the affinities of these interactions, which regulate host cell infection, were quantified. Together, these findings highlight the tremendous potential of SLOMO-nMS to accelerate the characterization of protein assembly pathways and thermodynamics and, in so doing, enhance fundamental biological understanding and facilitate therapeutic development. https://orcid.org/0000-0002-3389-7112.
Collapse
Affiliation(s)
- Duong T Bui
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2G2
| | - Elena N Kitova
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2G2
| | - Pavel I Kitov
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2G2
| | - Ling Han
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2G2
| | - Lara K Mahal
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2G2
| | - John S Klassen
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2G2
| |
Collapse
|
9
|
Han C, Zhang D, Xia S, Zhang Y. Accurate Prediction of NMR Chemical Shifts: Integrating DFT Calculations with Three-Dimensional Graph Neural Networks. J Chem Theory Comput 2024; 20:5250-5258. [PMID: 38842505 PMCID: PMC11209944 DOI: 10.1021/acs.jctc.4c00422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/25/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024]
Abstract
Computer prediction of NMR chemical shifts plays an increasingly important role in molecular structure assignment and elucidation for organic molecule studies. Density functional theory (DFT) and gauge-including atomic orbital (GIAO) have established a framework to predict NMR chemical shifts but often at a significant computational expense with a limited prediction accuracy. Recent advancements in deep learning methods, especially graph neural networks (GNNs), have shown promise in improving the accuracy of predicting experimental chemical shifts, either by using 2D molecular topological features or 3D conformational representation. This study presents a new 3D GNN model to predict 1H and 13C chemical shifts, CSTShift, that combines atomic features with DFT-calculated shielding tensor descriptors, capturing both isotropic and anisotropic shielding effects. Utilizing the NMRShiftDB2 data set and conducting DFT optimization and GIAO calculations at the B3LYP/6-31G(d) level, we prepared the NMRShiftDB2-DFT data set of high-quality 3D structures and shielding tensors with corresponding experimentally measured 1H and 13C chemical shifts. The developed CSTShift models achieve the state-of-the-art prediction performance on both the NMRShiftDB2-DFT test data set and external CHESHIRE data set. Further case studies on identifying correct structures from two groups of constitutional isomers show its capability for structure assignment and elucidation. The source code and data are accessible at https://yzhang.hpc.nyu.edu/IMA.
Collapse
Affiliation(s)
- Chao Han
- Department
of Chemistry, New York University, New York, New York 10003, United States
| | - Dongdong Zhang
- Department
of Chemistry, New York University, New York, New York 10003, United States
| | - Song Xia
- Department
of Chemistry, New York University, New York, New York 10003, United States
| | - Yingkai Zhang
- Department
of Chemistry, New York University, New York, New York 10003, United States
- Simons
Center for Computational Physical Chemistry at New York University, New York, New York 10003, United States
- NYU-ECNU
Center for Computational Chemistry at NYU Shanghai, Shanghai 200062, China
| |
Collapse
|
10
|
Li M, Tian X, Du W, Wang X, Lei J, Gao T, Zou S, Xu X, Wang H, Chen J, Gou Q. Decrypting the critical point of internal rotation of formaldehyde: A rotational study of the acrolein-formaldehyde complex. J Chem Phys 2024; 160:234303. [PMID: 38884398 DOI: 10.1063/5.0211674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 05/31/2024] [Indexed: 06/18/2024] Open
Abstract
The rotational spectrum of an acrolein-formaldehyde complex has been characterized using pulsed jet Fourier transform microwave spectroscopy complemented with quantum chemical calculations. One isomer has been observed in pulsed jets, which is stabilized by a dominant O=C⋯O tetrel bond (n → π* interaction) and a secondary C-H⋯O hydrogen bond. Splittings arising from the internal rotation of formaldehyde around its C2v axis were also observed, from which its V2 barrier was evaluated. It seems that when V2 equals or exceeds 4.61 kJ mol-1, no splitting of the spectral lines of the rotational spectrum was observed. The nature of the non-covalent interactions of the target complex is elucidated through natural bond orbital analysis. These findings contribute to a deeper understanding on the non-covalent interactions within the dimeric complex formed by two aldehydes.
Collapse
Affiliation(s)
- Meiyue Li
- School of Chemistry and Chemical Engineering, Chongqing University, Daxuecheng South Rd. 55, 401331 Chongqing, China
| | - Xiao Tian
- School of Chemistry and Chemical Engineering, Chongqing University, Daxuecheng South Rd. 55, 401331 Chongqing, China
| | - Weiping Du
- School of Chemistry and Chemical Engineering, Chongqing University, Daxuecheng South Rd. 55, 401331 Chongqing, China
| | - Xiujuan Wang
- School of Chemistry and Chemical Engineering, Chongqing University, Daxuecheng South Rd. 55, 401331 Chongqing, China
| | - Juncheng Lei
- School of Chemistry and Chemical Engineering, Chongqing University, Daxuecheng South Rd. 55, 401331 Chongqing, China
| | - Tianyue Gao
- School of Chemistry and Chemical Engineering, Chongqing University, Daxuecheng South Rd. 55, 401331 Chongqing, China
| | - Siyu Zou
- School of Chemistry and Chemical Engineering, Chongqing University, Daxuecheng South Rd. 55, 401331 Chongqing, China
| | - Xuefang Xu
- School of Chemistry and Chemical Engineering, Chongqing University, Daxuecheng South Rd. 55, 401331 Chongqing, China
| | - Hao Wang
- State Key Laboratory of Coal Conversion, Institute of Coal Chemistry, Chinese Academy of Sciences, Taoyuan South Rd. 27, Taiyuan 030001, Shanxi, China
| | - Junhua Chen
- School of Pharmacy, Guizhou Medical University, Guiyang 561113, Guizhou, China
| | - Qian Gou
- School of Chemistry and Chemical Engineering, Chongqing University, Daxuecheng South Rd. 55, 401331 Chongqing, China
- School of Chemistry and Chemical Engineering, Chongqing Key Laboratory of Chemical Theory and Mechanism, Chongqing University, Daxuecheng South Rd. 55, 401331 Chongqing, China
| |
Collapse
|
11
|
Vitale GA, Geibel C, Minda V, Wang M, Aron AT, Petras D. Connecting metabolome and phenotype: recent advances in functional metabolomics tools for the identification of bioactive natural products. Nat Prod Rep 2024; 41:885-904. [PMID: 38351834 PMCID: PMC11186733 DOI: 10.1039/d3np00050h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Indexed: 06/20/2024]
Abstract
Covering: 1995 to 2023Advances in bioanalytical methods, particularly mass spectrometry, have provided valuable molecular insights into the mechanisms of life. Non-targeted metabolomics aims to detect and (relatively) quantify all observable small molecules present in a biological system. By comparing small molecule abundances between different conditions or timepoints in a biological system, researchers can generate new hypotheses and begin to understand causes of observed phenotypes. Functional metabolomics aims to investigate the functional roles of metabolites at the scale of the metabolome. However, most functional metabolomics studies rely on indirect measurements and correlation analyses, which leads to ambiguity in the precise definition of functional metabolomics. In contrast, the field of natural products has a history of identifying the structures and bioactivities of primary and specialized metabolites. Here, we propose to expand and reframe functional metabolomics by integrating concepts from the fields of natural products and chemical biology. We highlight emerging functional metabolomics approaches that shift the focus from correlation to physical interactions, and we discuss how this allows researchers to uncover causal relationships between molecules and phenotypes.
Collapse
Affiliation(s)
- Giovanni Andrea Vitale
- CMFI Cluster of Excellence, Interfaculty Institute of Microbiology and Medicine, University of Tuebingen, Tuebingen, Germany
| | - Christian Geibel
- CMFI Cluster of Excellence, Interfaculty Institute of Microbiology and Medicine, University of Tuebingen, Tuebingen, Germany
| | - Vidit Minda
- Division of Pharmacology and Pharmaceutical Sciences, University of Missouri - Kansas City, Kansas City, USA
- Department of Chemistry and Biochemistry, University of Denver, Denver, USA.
| | - Mingxun Wang
- Department of Computer Science, University of California Riverside, Riverside, USA.
| | - Allegra T Aron
- Department of Chemistry and Biochemistry, University of Denver, Denver, USA.
| | - Daniel Petras
- CMFI Cluster of Excellence, Interfaculty Institute of Microbiology and Medicine, University of Tuebingen, Tuebingen, Germany
- Department of Biochemistry, University of California Riverside, Riverside, USA.
| |
Collapse
|
12
|
Štěpánová S, Kašička V. Determination of physicochemical parameters of (bio)molecules and (bio)particles by capillary electromigration methods. J Sep Sci 2024; 47:e2400174. [PMID: 38867483 DOI: 10.1002/jssc.202400174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/03/2024] [Accepted: 05/10/2024] [Indexed: 06/14/2024]
Abstract
The review provides an overview of recent developments and applications of capillary electromigration (CE) methods for the determination of important physicochemical parameters of various (bio)molecules and (bio)particles. These parameters include actual and limiting (absolute) ionic mobilities, effective electrophoretic mobilities, effective charges, isoelectric points, electrokinetic potentials, hydrodynamic radii, diffusion coefficients, relative molecular masses, acidity (ionization) constants, binding constants and stoichiometry of (bio)molecular complexes, changes of Gibbs free energy, enthalpy and entropy and rate constants of chemical reactions and interactions, retention factors and partition and distribution coefficients. For the determination of these parameters, the following CE methods are employed: zone electrophoresis in a free solution or in sieving media, isotachophoresis, isoelectric focusing, affinity electrophoresis, electrokinetic chromatography, and electrochromatography. In the individual sections, the procedures for the determination of the above parameters by the particular CE methods are described.
Collapse
Affiliation(s)
- Sille Štěpánová
- Electromigration methods, Institute of Organic Chemistry and Biochemistry, The Czech Academy of Sciences, Prague, Czech Republic
| | - Václav Kašička
- Electromigration methods, Institute of Organic Chemistry and Biochemistry, The Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
13
|
Noble M, Colussi DM, Junop M, Stathopulos PB. The MCU and MCUb amino-terminal domains tightly interact: mechanisms for low conductance assembly of the mitochondrial calcium uniporter complex. iScience 2024; 27:109699. [PMID: 38706857 PMCID: PMC11068563 DOI: 10.1016/j.isci.2024.109699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 02/12/2024] [Accepted: 04/05/2024] [Indexed: 05/07/2024] Open
Abstract
The mitochondrial calcium (Ca2+) uniporter (MCU) complex is regulated via integration of the MCU dominant negative beta subunit (MCUb), a low conductance paralog of the main MCU pore forming protein. The MCU amino (N)-terminal domain (NTD) also modulates channel function through cation binding to the MCU regulating acidic patch (MRAP). MCU and MCUb have high sequence similarities, yet the structural and functional roles of MCUb-NTD remain unknown. Here, we report that MCUb-NTD exhibits α-helix/β-sheet structure with a high thermal stability, dependent on protein concentration. Remarkably, MCU- and MCUb-NTDs heteromerically interact with ∼nM affinity, increasing secondary structure and stability and structurally perturbing MRAP. Further, we demonstrate MCU and MCUb co-localization is suppressed upon NTD deletion concomitant with increased mitochondrial Ca2+ uptake. Collectively, our data show that MCU:MCUb NTD tight interactions are promoted by enhanced regular structure and stability, augmenting MCU:MCUb co-localization, lowering mitochondrial Ca2+ uptake and implicating an MRAP-sensing mechanism.
Collapse
Affiliation(s)
- Megan Noble
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A5C1, Canada
| | - Danielle M. Colussi
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A5C1, Canada
| | - Murray Junop
- Department of Biochemistry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A5C1, Canada
| | - Peter B. Stathopulos
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A5C1, Canada
| |
Collapse
|
14
|
Bahena Culhuac E, Bello M. Unveiling the Mechanisms of EGCG-p53 Interactions through Molecular Dynamics Simulations. ACS OMEGA 2024; 9:20066-20085. [PMID: 38737068 PMCID: PMC11080030 DOI: 10.1021/acsomega.3c10523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 03/24/2024] [Accepted: 03/27/2024] [Indexed: 05/14/2024]
Abstract
Green tea consumption is associated with protective and preventive effects against various types of cancer. These effects are attributed to polyphenols, particularly epigallocatechin-3-gallate (EGCG). EGCG acts by directly inhibiting tumor suppressor protein p53. The binding mechanism by which EGCG inhibits p53 activity is associated with residues Trp23-Lys24 and Pro47-Thr55 within the p53 N-terminal domain (NTD). However, the structural and thermodynamic aspects of the interaction between EGCG and p53 are poorly understood. Therefore, based on crystallographic data, we combine docking, molecular dynamics (MD) simulations, and molecular mechanics generalized Born surface area approaches to explore the intricacies of the EGCG-p53 binding mechanism. A triplicate microsecond MD simulation for each system is initially performed to capture diverse p53 NTD conformations. From the start, the most populated cluster of the second run (R2-1) stands out due to a unique opening between Trp23 and Trp53. During MD simulations, this conformation allows EGCG to sustain a high level of stability and affinity while interacting with both regions of interest and deepening the binding pocket. Structural analysis emphasizes the significance of pyrogallol motifs in EGCG binding. Therefore, the conformational shift in this gap is pivotal, enabling EGCG to impede p53 interactions and manifest its anticancer properties. These findings enhance the present comprehension of the anticancer properties of green tea polyphenols and pave the way for future therapeutic developments.
Collapse
Affiliation(s)
- Erick Bahena Culhuac
- Laboratorio
de Diseño y Desarrollo de Nuevos Fármacos e Innovación
Biotecnológica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
- Universidad
Autónoma del Estado de México Facultad de Ciencias, Toluca 50000, Mexico
| | - Martiniano Bello
- Laboratorio
de Diseño y Desarrollo de Nuevos Fármacos e Innovación
Biotecnológica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
| |
Collapse
|
15
|
Stadler KA, Ortiz-Joya LJ, Singh Sahrawat A, Buhlheller C, Gruber K, Pavkov-Keller T, O'Hagan TB, Guarné A, Pulido S, Marín-Villa M, Zangger K, Gubensäk N. Structural investigation of Trypanosoma cruzi Akt-like kinase as drug target against Chagas disease. Sci Rep 2024; 14:10039. [PMID: 38693166 PMCID: PMC11063076 DOI: 10.1038/s41598-024-59654-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/12/2024] [Indexed: 05/03/2024] Open
Abstract
According to the World Health Organization, Chagas disease (CD) is the most prevalent poverty-promoting neglected tropical disease. Alarmingly, climate change is accelerating the geographical spreading of CD causative parasite, Trypanosoma cruzi, which additionally increases infection rates. Still, CD treatment remains challenging due to a lack of safe and efficient drugs. In this work, we analyze the viability of T. cruzi Akt-like kinase (TcAkt) as drug target against CD including primary structural and functional information about a parasitic Akt protein. Nuclear Magnetic Resonance derived information in combination with Molecular Dynamics simulations offer detailed insights into structural properties of the pleckstrin homology (PH) domain of TcAkt and its binding to phosphatidylinositol phosphate ligands (PIP). Experimental data combined with Alpha Fold proposes a model for the mechanism of action of TcAkt involving a PIP-induced disruption of the intramolecular interface between the kinase and the PH domain resulting in an open conformation enabling TcAkt kinase activity. Further docking experiments reveal that TcAkt is recognized by human inhibitors PIT-1 and capivasertib, and TcAkt inhibition by UBMC-4 and UBMC-6 is achieved via binding to TcAkt kinase domain. Our in-depth structural analysis of TcAkt reveals potential sites for drug development against CD, located at activity essential regions.
Collapse
Affiliation(s)
- Karina A Stadler
- Institute of Chemistry/Organic and Bioorganic Chemistry, University of Graz, Graz, Austria
| | - Lesly J Ortiz-Joya
- Institute of Chemistry/Organic and Bioorganic Chemistry, University of Graz, Graz, Austria
- Programa de Estudio y Control de Enfermedades Tropicales (PECET), Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
- Department of Biochemistry, McGill University, Montreal, Canada
| | - Amit Singh Sahrawat
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- Innophore GmbH, Graz, Austria
| | | | - Karl Gruber
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- Innophore GmbH, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Tea Pavkov-Keller
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | | | - Alba Guarné
- Department of Biochemistry, McGill University, Montreal, Canada
| | - Sergio Pulido
- Programa de Estudio y Control de Enfermedades Tropicales (PECET), Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
- LifeFactors ZF SAS, Rionegro, Colombia
| | - Marcel Marín-Villa
- Programa de Estudio y Control de Enfermedades Tropicales (PECET), Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Klaus Zangger
- Institute of Chemistry/Organic and Bioorganic Chemistry, University of Graz, Graz, Austria.
- Field of Excellence BioHealth, University of Graz, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
| | - Nina Gubensäk
- Institute of Molecular Biosciences, University of Graz, Graz, Austria.
| |
Collapse
|
16
|
Lu B, Liao SM, Liang SJ, Peng LX, Li JX, Liu XH, Huang RB, Zhou GP. The Bifunctional Effects of Lactoferrin (LFcinB11) in Inhibiting Neural Cell Adhesive Molecule (NCAM) Polysialylation and the Release of Neutrophil Extracellular Traps (NETs). Int J Mol Sci 2024; 25:4641. [PMID: 38731861 PMCID: PMC11083048 DOI: 10.3390/ijms25094641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/17/2024] [Accepted: 04/19/2024] [Indexed: 05/13/2024] Open
Abstract
The expression of polysialic acid (polySia) on the neuronal cell adhesion molecule (NCAM) is called NCAM-polysialylation, which is strongly related to the migration and invasion of tumor cells and aggressive clinical status. Thus, it is important to select a proper drug to block tumor cell migration during clinical treatment. In this study, we proposed that lactoferrin (LFcinB11) may be a better candidate for inhibiting NCAM polysialylation when compared with CMP and low-molecular-weight heparin (LMWH), which were determined based on our NMR studies. Furthermore, neutrophil extracellular traps (NETs) represent the most dramatic stage in the cell death process, and the release of NETs is related to the pathogenesis of autoimmune and inflammatory disorders, with proposed involvement in glomerulonephritis, chronic lung disease, sepsis, and vascular disorders. In this study, the molecular mechanisms involved in the inhibition of NET release using LFcinB11 as an inhibitor were also determined. Based on these results, LFcinB11 is proposed as being a bifunctional inhibitor for inhibiting both NCAM polysialylation and the release of NETs.
Collapse
Affiliation(s)
- Bo Lu
- National Key Laboratory of Non-Food Biomass Energy Technology, National Engineering Research Center for Non-Food Biorefinery, Institute of Biological Science and Technology, Guangxi Academy of Sciences, 98 Daling Road, Nanning 530007, China; (B.L.); (S.-M.L.); (S.-J.L.); (L.-X.P.); (J.-X.L.)
| | - Si-Ming Liao
- National Key Laboratory of Non-Food Biomass Energy Technology, National Engineering Research Center for Non-Food Biorefinery, Institute of Biological Science and Technology, Guangxi Academy of Sciences, 98 Daling Road, Nanning 530007, China; (B.L.); (S.-M.L.); (S.-J.L.); (L.-X.P.); (J.-X.L.)
| | - Shi-Jie Liang
- National Key Laboratory of Non-Food Biomass Energy Technology, National Engineering Research Center for Non-Food Biorefinery, Institute of Biological Science and Technology, Guangxi Academy of Sciences, 98 Daling Road, Nanning 530007, China; (B.L.); (S.-M.L.); (S.-J.L.); (L.-X.P.); (J.-X.L.)
| | - Li-Xin Peng
- National Key Laboratory of Non-Food Biomass Energy Technology, National Engineering Research Center for Non-Food Biorefinery, Institute of Biological Science and Technology, Guangxi Academy of Sciences, 98 Daling Road, Nanning 530007, China; (B.L.); (S.-M.L.); (S.-J.L.); (L.-X.P.); (J.-X.L.)
| | - Jian-Xiu Li
- National Key Laboratory of Non-Food Biomass Energy Technology, National Engineering Research Center for Non-Food Biorefinery, Institute of Biological Science and Technology, Guangxi Academy of Sciences, 98 Daling Road, Nanning 530007, China; (B.L.); (S.-M.L.); (S.-J.L.); (L.-X.P.); (J.-X.L.)
| | - Xue-Hui Liu
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China;
| | - Ri-Bo Huang
- National Key Laboratory of Non-Food Biomass Energy Technology, National Engineering Research Center for Non-Food Biorefinery, Institute of Biological Science and Technology, Guangxi Academy of Sciences, 98 Daling Road, Nanning 530007, China; (B.L.); (S.-M.L.); (S.-J.L.); (L.-X.P.); (J.-X.L.)
- Rocky Mount Life Sciences Institute, Rocky Mount, NC 27804, USA
| | - Guo-Ping Zhou
- National Key Laboratory of Non-Food Biomass Energy Technology, National Engineering Research Center for Non-Food Biorefinery, Institute of Biological Science and Technology, Guangxi Academy of Sciences, 98 Daling Road, Nanning 530007, China; (B.L.); (S.-M.L.); (S.-J.L.); (L.-X.P.); (J.-X.L.)
- Rocky Mount Life Sciences Institute, Rocky Mount, NC 27804, USA
| |
Collapse
|
17
|
Toscano G, Höfurthner T, Nagl B, Beier A, Mayer M, Geist L, McConnell DB, Weinstabl H, Konrat R, Lichtenecker RJ. 13 Cβ-Valine and 13 Cγ-Leucine Methine Labeling To Probe Protein Ligand Interaction. Chembiochem 2024; 25:e202300762. [PMID: 38294275 DOI: 10.1002/cbic.202300762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 02/01/2024]
Abstract
Precise information regarding the interaction between proteins and ligands at molecular resolution is crucial for effectively guiding the optimization process from initial hits to lead compounds in early stages of drug development. In this study, we introduce a novel aliphatic side chain isotope-labeling scheme to directly probe interactions between ligands and aliphatic sidechains using NMR techniques. To demonstrate the applicability of this method, we selected a set of Brd4-BD1 binders and analyzed 1 H chemical shift perturbation resulting from CH-π interaction of Hβ -Val and Hγ -Leu as CH donors with corresponding ligand aromatic moieties as π acceptors.
Collapse
Affiliation(s)
- Giorgia Toscano
- Christian Doppler Laboratory for High-Content Structural Biology and Biotechnology, Institute of Organic Chemistry, University of Vienna, Währingerstraße 38, 1090, Vienna, Austria
- Vienna Doctoral School of Chemistry, University of Vienna, Währingerstr. 38, 1090, Vienna, Austria
| | - Theresa Höfurthner
- Christian Doppler Laboratory for High-Content Structural Biology and Biotechnology, Max Perutz Laboratories, Department of Structural and Computational Biology, Campus Vienna Biocenter 5, 1030, Vienna, Austria
- Vienna Doctoral School of Chemistry, University of Vienna, Währingerstr. 38, 1090, Vienna, Austria
| | - Benjamin Nagl
- Christian Doppler Laboratory for High-Content Structural Biology and Biotechnology, Institute of Organic Chemistry, University of Vienna, Währingerstraße 38, 1090, Vienna, Austria
| | - Andreas Beier
- Christian Doppler Laboratory for High-Content Structural Biology and Biotechnology, Max Perutz Laboratories, Department of Structural and Computational Biology, Campus Vienna Biocenter 5, 1030, Vienna, Austria
| | - Moriz Mayer
- Boehringer Ingelheim RCV GmbH & Co. KG, Dr. Boehringer, Gasse 5-Wien, 11, 1121, Vienna
| | - Leonhard Geist
- Boehringer Ingelheim RCV GmbH & Co. KG, Dr. Boehringer, Gasse 5-Wien, 11, 1121, Vienna
| | - Darryl B McConnell
- Boehringer Ingelheim RCV GmbH & Co. KG, Dr. Boehringer, Gasse 5-Wien, 11, 1121, Vienna
| | - Harald Weinstabl
- Boehringer Ingelheim RCV GmbH & Co. KG, Dr. Boehringer, Gasse 5-Wien, 11, 1121, Vienna
| | - Robert Konrat
- Christian Doppler Laboratory for High-Content Structural Biology and Biotechnology, Max Perutz Laboratories, Department of Structural and Computational Biology, Campus Vienna Biocenter 5, 1030, Vienna, Austria
- MAG-LAB, Karl-Farkas Gasse 22, 1030, Vienna
| | - Roman J Lichtenecker
- Christian Doppler Laboratory for High-Content Structural Biology and Biotechnology, Institute of Organic Chemistry, University of Vienna, Währingerstraße 38, 1090, Vienna, Austria
- MAG-LAB, Karl-Farkas Gasse 22, 1030, Vienna
| |
Collapse
|
18
|
Hőgye F, Farkas LB, Balogh ÁK, Szilágyi L, Alnukari S, Bajza I, Borbás A, Fehér K, Illyés TZ, Timári I. Saturation Transfer Difference NMR and Molecular Docking Interaction Study of Aralkyl-Thiodigalactosides as Potential Inhibitors of the Human-Galectin-3 Protein. Int J Mol Sci 2024; 25:1742. [PMID: 38339036 PMCID: PMC10855533 DOI: 10.3390/ijms25031742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Human Galectin-3 (hGal-3) is a protein that selectively binds to β-galactosides and holds diverse roles in both normal and pathological circumstances. Therefore, targeting hGal-3 has become a vibrant area of research in the pharmaceutical chemistry. As a step towards the development of novel hGal-3 inhibitors, we synthesized and investigated derivatives of thiodigalactoside (TDG) modified with different aromatic substituents. Specifically, we describe a high-yielding synthetic route of thiodigalactoside (TDG); an optimized procedure for the synthesis of the novel 3,3'-di-O-(quinoline-2-yl)methyl)-TDG and three other known, symmetric 3,3'-di-O-TDG derivatives ((naphthalene-2yl)methyl, benzyl, (7-methoxy-2H-1-benzopyran-2-on-4-yl)methyl). In the present study, using competition Saturation Transfer Difference (STD) NMR spectroscopy, we determined the dissociation constant (Kd) of the former three TDG derivatives produced to characterize the strength of the interaction with the target protein (hGal-3). Based on the Kd values determined, the (naphthalen-2-yl)methyl, the (quinolin-2-yl)methyl and the benzyl derivatives bind to hGal-3 94, 30 and 24 times more strongly than TDG. Then, we studied the binding modes of the derivatives in silico by molecular docking calculations. Docking poses similar to the canonical binding modes of well-known hGal-3 inhibitors have been found. However, additional binding forces, cation-π interactions between the arginine residues in the binding pocket of the protein and the aromatic groups of the ligands, have been established as significant features. Our results offer a molecular-level understanding of the varying affinities observed among the synthesized thiodigalactoside derivatives, which can be a key aspect in the future development of more effective ligands of hGal-3.
Collapse
Affiliation(s)
- Fanni Hőgye
- Department of Organic Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (F.H.); (L.B.F.); (L.S.)
| | - László Bence Farkas
- Department of Organic Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (F.H.); (L.B.F.); (L.S.)
- HUN-REN-UD Molecular Recognition and Interaction Research Group, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (Á.K.B.); (S.A.); (A.B.); (K.F.)
| | - Álex Kálmán Balogh
- HUN-REN-UD Molecular Recognition and Interaction Research Group, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (Á.K.B.); (S.A.); (A.B.); (K.F.)
| | - László Szilágyi
- Department of Organic Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (F.H.); (L.B.F.); (L.S.)
| | - Samar Alnukari
- HUN-REN-UD Molecular Recognition and Interaction Research Group, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (Á.K.B.); (S.A.); (A.B.); (K.F.)
| | - István Bajza
- GlycOptim Kft., Egyetem tér 1, H-4032 Debrecen, Hungary;
| | - Anikó Borbás
- HUN-REN-UD Molecular Recognition and Interaction Research Group, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (Á.K.B.); (S.A.); (A.B.); (K.F.)
- Department of Pharmaceutical Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Krisztina Fehér
- HUN-REN-UD Molecular Recognition and Interaction Research Group, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (Á.K.B.); (S.A.); (A.B.); (K.F.)
| | - Tünde Zita Illyés
- Department of Organic Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (F.H.); (L.B.F.); (L.S.)
| | - István Timári
- Department of Organic Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (F.H.); (L.B.F.); (L.S.)
- HUN-REN-UD Molecular Recognition and Interaction Research Group, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (Á.K.B.); (S.A.); (A.B.); (K.F.)
| |
Collapse
|
19
|
Georges T, Chèvre R, Cousin SF, Gervais C, Thureau P, Mollica G, Azaïs T. 43Ca MAS-DNP NMR of Frozen Solutions for the Investigation of Calcium Ion Complexation. ACS OMEGA 2024; 9:4881-4891. [PMID: 38313477 PMCID: PMC10831850 DOI: 10.1021/acsomega.3c08292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/11/2023] [Accepted: 12/26/2023] [Indexed: 02/06/2024]
Abstract
Calcium ion complexation in aqueous solutions is of paramount importance in biology as it is related to cell signaling, muscle contraction, or biomineralization. However, Ca2+-complexes are dynamic soluble entities challenging to describe at the molecular level. Nuclear magnetic resonance appears as a method of choice to probe Ca2+-complexes. However, 43Ca NMR exhibits severe limitations arising from the low natural abundance coupled to the low gyromagnetic ratio and the quadrupolar nature of 43Ca, which overall make it a very unreceptive nucleus. Here, we show that 43Ca dynamic nuclear polarization (DNP) NMR of 43Ca-labeled frozen solutions is an efficient approach to enhance the NMR receptivity of 43Ca and to obtain structural insights about calcium ions complexed with representative ligands including water molecules, ethylenediaminetetraacetic acid (EDTA), and l-aspartic acid (l-Asp). In these conditions and in combination with numerical simulations and calculations, we show that 43Ca nuclei belonging to Ca2+ complexed to the investigated ligands exhibit rather low quadrupolar couplings (with CQ typically ranging from 0.6 to 1 MHz) due to high symmetrical environments and potential residual dynamics in vitrified solutions at a temperature of 100 K. As a consequence, when 1H→43Ca cross-polarization (CP) is used to observe 43Ca central transition, "high-power" νRF(43Ca) conditions, typically used to detect spin 1/2 nuclei, provide ∼120 times larger sensitivity than "low-power" conditions usually employed for detection of quadrupolar nuclei. These "high-power" CPMAS conditions allow two-dimensional (2D) 1H-43Ca HetCor spectra to be readily recorded, highlighting various Ca2+-ligand interactions in solution. This significant increase in 43Ca NMR sensitivity results from the combination of distinct advantages: (i) an efficient 1H-mediated polarization transfer from DNP, resembling the case of low-natural-abundance spin 1/2 nuclei, (ii) a reduced dynamics, allowing the use of CP as a sensitivity enhancement technique, and (iii) the presence of a relatively highly symmetrical Ca environment, which, combined to residual dynamics, leads to the averaging of the quadrupolar interaction and hence to efficient high-power CP conditions. Interestingly, these results indicate that the use of high-power CP conditions is an effective way of selecting symmetrical and/or dynamic 43Ca environments of calcium-containing frozen solution, capable of filtering out more rigid and/or anisotropic 43Ca sites characterized by larger quadrupolar constants. This approach could open the way to the atomic-level investigation of calcium environments in more complex, heterogeneous frozen solutions, such as those encountered at the early stages of calcium phosphate or calcium carbonate biomineralization events.
Collapse
Affiliation(s)
- Tristan Georges
- Sorbonne
Université, CNRS, Laboratoire de Chimie de la Matière
Condensée de Paris (LCMCP), 4 Place Jussieu, 75005 Paris, France
| | - Romain Chèvre
- Aix
Marseille Univ, CNRS, ICR, 13397 Marseille, France
| | | | - Christel Gervais
- Sorbonne
Université, CNRS, Laboratoire de Chimie de la Matière
Condensée de Paris (LCMCP), 4 Place Jussieu, 75005 Paris, France
| | | | | | - Thierry Azaïs
- Sorbonne
Université, CNRS, Laboratoire de Chimie de la Matière
Condensée de Paris (LCMCP), 4 Place Jussieu, 75005 Paris, France
| |
Collapse
|
20
|
Erba EB, Pastore A. The Complementarity of Nuclear Magnetic Resonance and Native Mass Spectrometry in Probing Protein-Protein Interactions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 3234:109-123. [PMID: 38507203 DOI: 10.1007/978-3-031-52193-5_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Nuclear magnetic resonance (NMR) and native mass spectrometry (MS) are mature physicochemical techniques with long histories and important applications. NMR spectroscopy provides detailed information about the structure, dynamics, interactions, and chemical environment of biomolecules. MS is an effective approach for determining the mass of biomolecules with high accuracy, sensitivity, and speed. The two techniques offer unique advantages and provide solid tools for structural biology. In the present review, we discuss their individual merits in the context of their applications to structural studies in biology with specific focus on protein interactions and evaluate their limitations. We provide specific examples in which these techniques can complement each other, providing new information on the same scientific case. We discuss how the field may develop and what challenges are expected in the future. Overall, the combination of NMR and MS plays an increasingly important role in integrative structural biology, assisting scientists in deciphering the three-dimensional structure of composite macromolecular assemblies.
Collapse
|
21
|
Voigt B, Bhatia T, Hesselbarth J, Baumann M, Schmidt C, Ott M, Balbach J. The Prenucleation Equilibrium of the Parathyroid Hormone Determines the Critical Aggregation Concentration and Amyloid Fibril Nucleation. Chemphyschem 2023; 24:e202300439. [PMID: 37477386 DOI: 10.1002/cphc.202300439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/18/2023] [Accepted: 07/18/2023] [Indexed: 07/22/2023]
Abstract
Nucleation and growth of amyloid fibrils were found to only occur in supersaturated solutions above a critical concentration (ccrit ). The biophysical meaning of ccrit remained mostly obscure, since typical low values of ccrit in the sub-μM range hamper investigations of potential oligomeric states and their structure. Here, we investigate the parathyroid hormone PTH84 as an example of a functional amyloid fibril forming peptide with a comparably high ccrit of 67±21 μM. We describe a complex concentration dependent prenucleation ensemble of oligomers of different sizes and secondary structure compositions and highlight the occurrence of a trimer and tetramer at ccrit as possible precursors for primary fibril nucleation. Furthermore, the soluble state found in equilibrium with fibrils adopts to the prenucleation state present at ccrit . Our study sheds light onto early events of amyloid formation directly related to the critical concentration and underlines oligomer formation as a key feature of fibril nucleation. Our results contribute to a deeper understanding of the determinants of supersaturated peptide solutions. In the current study we present a biophysical approach to investigate ccrit of amyloid fibril formation of PTH84 in terms of secondary structure, cluster size and residue resolved intermolecular interactions during oligomer formation. Throughout the investigated range of concentrations (1 μM to 500 μM) we found different states of oligomerization with varying ability to contribute to primary fibril nucleation and with a concentration dependent equilibrium. In this context, we identified the previously described ccrit of PTH84 to mark a minimum concentration for the formation of homo-trimers/tetramers. These investigations allowed us to characterize molecular interactions of various oligomeric states that are further converted into elongation competent fibril nuclei during the lag phase of a functional amyloid forming peptide.
Collapse
Affiliation(s)
- Bruno Voigt
- Martin Luther University Halle-Wittenberg, Institute of Physics, Betty-Heimann-Straße 7, 06120, Halle, Germany
| | - Twinkle Bhatia
- Martin Luther University Halle-Wittenberg, Institute of Biochemistry and Biotechnology, Kurt-Mothes-Straße 3, 06120, Halle, Germany
| | - Julia Hesselbarth
- present address: Johannes Gutenberg University Mainz, Institute of Chemistry - Biochemistry, Biocenter II, Hanns-Dieter-Hüsch-Weg 17, 55128, Mainz, Germany
- Martin Luther University Halle-Wittenberg, Interdisciplinary Research Center HALOmem, Institute of Biochemistry and Biotechnology, Kurt-Mothes-Straße 3a, 06120, Halle, Germany
| | - Monika Baumann
- Martin Luther University Halle-Wittenberg, Institute of Physics, Betty-Heimann-Straße 7, 06120, Halle, Germany
| | - Carla Schmidt
- present address: Johannes Gutenberg University Mainz, Institute of Chemistry - Biochemistry, Biocenter II, Hanns-Dieter-Hüsch-Weg 17, 55128, Mainz, Germany
- Martin Luther University Halle-Wittenberg, Interdisciplinary Research Center HALOmem, Institute of Biochemistry and Biotechnology, Kurt-Mothes-Straße 3a, 06120, Halle, Germany
| | - Maria Ott
- Martin Luther University Halle-Wittenberg, Institute of Biochemistry and Biotechnology, Kurt-Mothes-Straße 3, 06120, Halle, Germany
| | - Jochen Balbach
- Martin Luther University Halle-Wittenberg, Institute of Physics, Betty-Heimann-Straße 7, 06120, Halle, Germany
| |
Collapse
|
22
|
Greisen PJ, Yi L, Zhou R, Zhou J, Johansson E, Dong T, Liu H, Johnsen LB, Lund S, Svensson LA, Zhu H, Thomas N, Yang Z, Østergaard H. Computational design of N-linked glycans for high throughput epitope profiling. Protein Sci 2023; 32:e4726. [PMID: 37421602 PMCID: PMC10521239 DOI: 10.1002/pro.4726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/12/2023] [Accepted: 07/06/2023] [Indexed: 07/10/2023]
Abstract
Efficient identification of epitopes is crucial for drug discovery and design as it enables the selection of optimal epitopes, expansion of lead antibody diversity, and verification of binding interface. Although high-resolution low throughput methods like x-ray crystallography can determine epitopes or protein-protein interactions accurately, they are time-consuming and can only be applied to a limited number of complexes. To overcome these limitations, we have developed a rapid computational method that incorporates N-linked glycans to mask epitopes or protein interaction surfaces, thereby providing a mapping of these regions. Using human coagulation factor IXa (fIXa) as a model system, we computationally screened 158 positions and expressed 98 variants to test experimentally for epitope mapping. We were able to delineate epitopes rapidly and reliably through the insertion of N-linked glycans that efficiently disrupted binding in a site-selective manner. To validate the efficacy of our method, we conducted ELISA experiments and high-throughput yeast surface display assays. Furthermore, x-ray crystallography was employed to verify the results, thereby recapitulating through the method of N-linked glycans a coarse-grained mapping of the epitope.
Collapse
Affiliation(s)
| | - Li Yi
- Global Research TechnologiesNovo Nordisk A/SMaaloevDenmark
| | - Rong Zhou
- Discovery Technology China, Novo Nordisk Research CentreNovo Nordisk A/SBeijingChina
| | - Jian Zhou
- Discovery Technology China, Novo Nordisk Research CentreNovo Nordisk A/SBeijingChina
| | - Eva Johansson
- Global Research TechnologiesNovo Nordisk A/SMaaloevDenmark
| | - Tiantang Dong
- Discovery Technology China, Novo Nordisk Research CentreNovo Nordisk A/SBeijingChina
| | - Haimo Liu
- Discovery Technology China, Novo Nordisk Research CentreNovo Nordisk A/SBeijingChina
| | | | - Søren Lund
- Global Research TechnologiesNovo Nordisk A/SMaaloevDenmark
| | | | - Haisun Zhu
- Discovery Technology China, Novo Nordisk Research CentreNovo Nordisk A/SBeijingChina
| | - Nidhin Thomas
- Digital Science and InnovationNovo Nordisk A/SSeattleUSA
| | - Zhiru Yang
- Discovery Technology China, Novo Nordisk Research CentreNovo Nordisk A/SBeijingChina
| | | |
Collapse
|
23
|
Gubensäk N, Sagmeister T, Buhlheller C, Geronimo BD, Wagner GE, Petrowitsch L, Gräwert MA, Rotzinger M, Berger TMI, Schäfer J, Usón I, Reidl J, Sánchez-Murcia PA, Zangger K, Pavkov-Keller T. Vibrio cholerae's ToxRS bile sensing system. eLife 2023; 12:e88721. [PMID: 37768326 PMCID: PMC10624426 DOI: 10.7554/elife.88721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 09/27/2023] [Indexed: 09/29/2023] Open
Abstract
The seventh pandemic of the diarrheal cholera disease, which began in 1960, is caused by the Gram-negative bacterium Vibrio cholerae. Its environmental persistence provoking recurring sudden outbreaks is enabled by V. cholerae's rapid adaption to changing environments involving sensory proteins like ToxR and ToxS. Located at the inner membrane, ToxR and ToxS react to environmental stimuli like bile acid, thereby inducing survival strategies for example bile resistance and virulence regulation. The presented crystal structure of the sensory domains of ToxR and ToxS in combination with multiple bile acid interaction studies, reveals that a bile binding pocket of ToxS is only properly folded upon binding to ToxR. Our data proposes an interdependent functionality between ToxR transcriptional activity and ToxS sensory function. These findings support the previously suggested link between ToxRS and VtrAC-like co-component systems. Besides VtrAC, ToxRS is now the only experimentally determined structure within this recently defined superfamily, further emphasizing its significance. In-depth analysis of the ToxRS complex reveals its remarkable conservation across various Vibrio species, underlining the significance of conserved residues in the ToxS barrel and the more diverse ToxR sensory domain. Unravelling the intricate mechanisms governing ToxRS's environmental sensing capabilities, provides a promising tool for disruption of this vital interaction, ultimately inhibiting Vibrio's survival and virulence. Our findings hold far-reaching implications for all Vibrio strains that rely on the ToxRS system as a shared sensory cornerstone for adapting to their surroundings.
Collapse
Affiliation(s)
- Nina Gubensäk
- Institute of Molecular Biosciences, University of GrazGrazAustria
| | - Theo Sagmeister
- Institute of Molecular Biosciences, University of GrazGrazAustria
| | | | - Bruno Di Geronimo
- Laboratory of Computer-Aided Molecular Design, Division of Medicinal Chemistry, Otto-Loewi Research Center, Medical University of GrazGrazAustria
| | - Gabriel E Wagner
- Institute of Chemistry / Organic and Bioorganic Chemistry, Medical University of GrazGrazAustria
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of GrazGrazAustria
| | | | | | - Markus Rotzinger
- Institute of Chemistry / Organic and Bioorganic Chemistry, Medical University of GrazGrazAustria
| | | | | | - Isabel Usón
- Institute of Molecular Biology of BarcelonaBarcelonaSpain
- ICREA, Institució Catalana de Recerca i Estudis AvançatsBarcelonaSpain
| | - Joachim Reidl
- Institute of Molecular Biosciences, University of GrazGrazAustria
- BioHealth Field of Excellence, University of GrazGrazAustria
- BioTechMed-GrazGrazAustria
| | - Pedro A Sánchez-Murcia
- Laboratory of Computer-Aided Molecular Design, Division of Medicinal Chemistry, Otto-Loewi Research Center, Medical University of GrazGrazAustria
| | - Klaus Zangger
- Institute of Chemistry / Organic and Bioorganic Chemistry, Medical University of GrazGrazAustria
- BioHealth Field of Excellence, University of GrazGrazAustria
- BioTechMed-GrazGrazAustria
| | - Tea Pavkov-Keller
- Institute of Molecular Biosciences, University of GrazGrazAustria
- BioHealth Field of Excellence, University of GrazGrazAustria
- BioTechMed-GrazGrazAustria
| |
Collapse
|
24
|
Nydegger DT, Pujol-Giménez J, Kandasamy P, Vogt B, Hediger MA. Applications of the Microscale Thermophoresis Binding Assay in COVID-19 Research. Viruses 2023; 15:1432. [PMID: 37515120 PMCID: PMC10386446 DOI: 10.3390/v15071432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/16/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
As the COVID-19 pandemic progresses, new variants of SARS-CoV-2 continue to emerge. This underscores the need to develop optimized tools to study such variants, along with new coronaviruses that may arise in the future. Such tools will also be instrumental in the development of new antiviral drugs. Here, we introduce microscale thermophoresis (MST) as a reliable and versatile tool for coronavirus research, which we demonstrate through three different applications described in this report: (1) binding of the SARS-CoV-2 spike receptor binding domain (RBD) to peptides as a strategy to prevent virus entry, (2) binding of the RBD to the viral receptor ACE2, and (3) binding of the RBD to ACE2 in complex with the amino acid transporter SLC6A20/SIT1 or its allelic variant rs61731475 (p.Ile529Val). Our results demonstrate that MST is a highly precise approach to studying protein-protein and/or protein-ligand interactions in coronavirus research, making it an ideal tool for studying viral variants and developing antiviral agents. Moreover, as shown in our results, a unique advantage of the MST assay over other available binding assays is the ability to measure interactions with membrane proteins in their near-native plasma membrane environment.
Collapse
Affiliation(s)
- Damian T Nydegger
- Department of Nephrology and Hypertension, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland
- Department of Biomedical Research, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland
| | - Jonai Pujol-Giménez
- Department of Nephrology and Hypertension, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland
- Department of Biomedical Research, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland
| | - Palanivel Kandasamy
- Department of Nephrology and Hypertension, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland
- Department of Biomedical Research, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland
| | - Bruno Vogt
- Department of Nephrology and Hypertension, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland
- Department of Biomedical Research, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland
| | - Matthias A Hediger
- Department of Nephrology and Hypertension, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland
- Department of Biomedical Research, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland
| |
Collapse
|
25
|
Malec K, Monaco S, Delso I, Nestorowicz J, Kozakiewicz-Latała M, Karolewicz B, Khimyak YZ, Angulo J, Nartowski KP. Unravelling the mechanisms of drugs partitioning phenomena in micellar systems via NMR spectroscopy. J Colloid Interface Sci 2023; 638:135-148. [PMID: 36736115 DOI: 10.1016/j.jcis.2023.01.063] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/12/2023] [Indexed: 01/22/2023]
Abstract
Despite extensive use of micelles in materials and colloidal science, their supramolecular organization as well as host-guest interactions within these dynamic assemblies are poorly understood. Small guest molecules in the presence of micelles undergo constant exchange between a micellar aggregate and the surrounding solution, posing a considerable challenge for their molecular level characterisation. In this work we reveal the interaction maps between small guest molecules and surfactants forming micelles via novel applications of NMR techniques supported with state-of-the-art analytical methods used in colloidal science. Model micelles composed of structurally distinct surfactants (block non-ionic polymer Pluronic® F-127, non-ionic surfactant Tween 20 or Tween 80, and ionic surfactant sodium lauryl sulphate, SLS) were selected and loaded with model small molecules of biological relevance (i.e. the drugs fluconazole, FLU or indomethacin, IMC) known to have different partition coefficients. Molecular level organization of FLU or IMC within hydrophilic and hydrophobic domains of micellar aggregates was established using the combination of NMR methods (1D 1H NMR, 1D 19F NMR, 2D 1H-1H NOESY and 2D 1H-19F HOESY, and the multifrequency-STD NMR) and corroborated with molecular dynamics (MD) simulations. This is the first application of multifrequency-STD NMR to colloidal systems, enabling us to elucidate intricately detailed patterns of drug/micelle interactions in a single NMR experiment within minutes. Importantly, our results indicate that flexible surfactants, such as block copolymers and polysorbates, form micellar aggregates with a surface composed of both hydrophilic and hydrophobic domains and do not follow the classical core-shell model of the micelle. We propose that the magnitude of changes in 1H chemical shifts corroborated with interaction maps obtained from DEEP-STD NMR and 2D NMR experiments can be used as an indicator of the strength of the guest-surfactant interactions. This NMR toolbox can be adopted for the analysis of broad range of colloidal host-guest systems from soft materials to biological systems.
Collapse
Affiliation(s)
- Katarzyna Malec
- Department of Drug Form Technology, Faculty of Pharmacy, Wroclaw Medical University, 211a Borowska Str, 50-556 Wroclaw, Poland
| | - Serena Monaco
- School of Pharmacy, University of East Anglia, Chancellors Drive, NR4 7TJ Norwich, UK
| | - Ignacio Delso
- School of Pharmacy, University of East Anglia, Chancellors Drive, NR4 7TJ Norwich, UK
| | - Justyna Nestorowicz
- Department of Drug Form Technology, Faculty of Pharmacy, Wroclaw Medical University, 211a Borowska Str, 50-556 Wroclaw, Poland
| | - Marta Kozakiewicz-Latała
- Department of Drug Form Technology, Faculty of Pharmacy, Wroclaw Medical University, 211a Borowska Str, 50-556 Wroclaw, Poland
| | - Bożena Karolewicz
- Department of Drug Form Technology, Faculty of Pharmacy, Wroclaw Medical University, 211a Borowska Str, 50-556 Wroclaw, Poland
| | - Yaroslav Z Khimyak
- School of Pharmacy, University of East Anglia, Chancellors Drive, NR4 7TJ Norwich, UK.
| | - Jesús Angulo
- School of Pharmacy, University of East Anglia, Chancellors Drive, NR4 7TJ Norwich, UK; Instituto de Investigaciones Químicas (CSIC-US), Avda. Américo Vespucio, 49, Sevilla 41092, Spain.
| | - Karol P Nartowski
- Department of Drug Form Technology, Faculty of Pharmacy, Wroclaw Medical University, 211a Borowska Str, 50-556 Wroclaw, Poland; School of Pharmacy, University of East Anglia, Chancellors Drive, NR4 7TJ Norwich, UK.
| |
Collapse
|
26
|
Mons E, Kim RQ, Mulder MPC. Technologies for Direct Detection of Covalent Protein-Drug Adducts. Pharmaceuticals (Basel) 2023; 16:547. [PMID: 37111304 PMCID: PMC10146396 DOI: 10.3390/ph16040547] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/29/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023] Open
Abstract
In the past two decades, drug candidates with a covalent binding mode have gained the interest of medicinal chemists, as several covalent anticancer drugs have successfully reached the clinic. As a covalent binding mode changes the relevant parameters to rank inhibitor potency and investigate structure-activity relationship (SAR), it is important to gather experimental evidence on the existence of a covalent protein-drug adduct. In this work, we review established methods and technologies for the direct detection of a covalent protein-drug adduct, illustrated with examples from (recent) drug development endeavors. These technologies include subjecting covalent drug candidates to mass spectrometric (MS) analysis, protein crystallography, or monitoring intrinsic spectroscopic properties of the ligand upon covalent adduct formation. Alternatively, chemical modification of the covalent ligand is required to detect covalent adducts by NMR analysis or activity-based protein profiling (ABPP). Some techniques are more informative than others and can also elucidate the modified amino acid residue or bond layout. We will discuss the compatibility of these techniques with reversible covalent binding modes and the possibilities to evaluate reversibility or obtain kinetic parameters. Finally, we expand upon current challenges and future applications. Overall, these analytical techniques present an integral part of covalent drug development in this exciting new era of drug discovery.
Collapse
Affiliation(s)
- Elma Mons
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (E.M.)
- Institute of Biology Leiden, Leiden University, 2333 BE Leiden, The Netherlands
| | - Robbert Q. Kim
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (E.M.)
| | - Monique P. C. Mulder
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (E.M.)
| |
Collapse
|
27
|
Fiorentino F, Rotili D, Mai A. Native mass spectrometry-directed drug discovery: Recent advances in investigating protein function and modulation. Drug Discov Today 2023; 28:103548. [PMID: 36871843 DOI: 10.1016/j.drudis.2023.103548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 02/15/2023] [Accepted: 02/28/2023] [Indexed: 03/07/2023]
Abstract
Native mass spectrometry (nMS) is a biophysical method for studying protein complexes and can provide insights into subunit stoichiometry and composition, protein-ligand, and protein-protein interactions (PPIs). These analyses are made possible by preserving non-covalent interactions in the gas phase, thereby allowing the analysis of proteins in their native state. Consequently, nMS has been increasingly applied in early drug discovery campaigns for the characterization of protein-drug interactions and the evaluation of PPI modulators. Here, we discuss recent developments in nMS-directed drug discovery and provide a timely perspective on the possible applications of this technology in drug discovery.
Collapse
Affiliation(s)
- Francesco Fiorentino
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | - Dante Rotili
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | - Antonello Mai
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; Pasteur Institute, Cenci-Bolognetti Foundation, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| |
Collapse
|
28
|
Kurbatov I, Dolgalev G, Arzumanian V, Kiseleva O, Poverennaya E. The Knowns and Unknowns in Protein-Metabolite Interactions. Int J Mol Sci 2023; 24:4155. [PMID: 36835565 PMCID: PMC9964805 DOI: 10.3390/ijms24044155] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/11/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Increasing attention has been focused on the study of protein-metabolite interactions (PMI), which play a key role in regulating protein functions and directing an orchestra of cellular processes. The investigation of PMIs is complicated by the fact that many such interactions are extremely short-lived, which requires very high resolution in order to detect them. As in the case of protein-protein interactions, protein-metabolite interactions are still not clearly defined. Existing assays for detecting protein-metabolite interactions have an additional limitation in the form of a limited capacity to identify interacting metabolites. Thus, although recent advances in mass spectrometry allow the routine identification and quantification of thousands of proteins and metabolites today, they still need to be improved to provide a complete inventory of biological molecules, as well as all interactions between them. Multiomic studies aimed at deciphering the implementation of genetic information often end with the analysis of changes in metabolic pathways, as they constitute one of the most informative phenotypic layers. In this approach, the quantity and quality of knowledge about PMIs become vital to establishing the full scope of crosstalk between the proteome and the metabolome in a biological object of interest. In this review, we analyze the current state of investigation into the detection and annotation of protein-metabolite interactions, describe the recent progress in developing associated research methods, and attempt to deconstruct the very term "interaction" to advance the field of interactomics further.
Collapse
Affiliation(s)
| | | | | | - Olga Kiseleva
- Institute of Biomedical Chemistry, Moscow 119121, Russia
| | | |
Collapse
|
29
|
NMR Analysis of the Interactions and Conformational Plasticity of Dynein Intermediate Chain. Methods Mol Biol 2023; 2623:241-256. [PMID: 36602690 DOI: 10.1007/978-1-0716-2958-1_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cytoplasmic dynein complexes play crucial roles in intracellular transport of cellular organelles. While the motor domain of dynein is well characterized by techniques such as X-ray crystallography and cryo-electron microscopy (Cryo-EM), structural representations of dynein usually include only the more packed and easily resolved regions and omit the long flexible and poorly structured regions. One such flexible region is the N-terminal half of the intermediate chain (IC), which contains almost 300 amino acids that are predicted to be disordered. This level of disorder makes IC impossible to study by X-ray crystallography and Cryo-EM, but amenable to study by solution nuclear magnetic resonance (NMR), a powerful technique that can elucidate residue-specific information in a dynamic ensemble of structures, and transient binding interactions of associated proteins. Here, we describe the methods we use to characterize flexible and disordered proteins including protein expression, purification, sample preparation, and NMR data acquisition and analysis.
Collapse
|
30
|
Thalhammer A, Bröker NK. Biophysical Approaches for the Characterization of Protein-Metabolite Interactions. Methods Mol Biol 2023; 2554:199-229. [PMID: 36178628 DOI: 10.1007/978-1-0716-2624-5_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
With an estimate of hundred thousands of protein molecules per cell and the number of metabolites several orders of magnitude higher, protein-metabolite interactions are omnipresent. In vitro analyses are one of the main pillars on the way to establish a solid understanding of how these interactions contribute to maintaining cellular homeostasis. A repertoire of biophysical techniques is available by which protein-metabolite interactions can be quantitatively characterized in terms of affinity, specificity, and kinetics in a broad variety of solution environments. Several of those provide information on local or global conformational changes of the protein partner in response to ligand binding. This review chapter gives an overview of the state-of-the-art biophysical toolbox for the study of protein-metabolite interactions. It briefly introduces basic principles, highlights recent examples from the literature, and pinpoints promising future directions.
Collapse
Affiliation(s)
- Anja Thalhammer
- Physical Biochemistry, University of Potsdam, Potsdam, Germany.
| | - Nina K Bröker
- Physical Biochemistry, University of Potsdam, Potsdam, Germany
- Health and Medical University Potsdam, Potsdam, Germany
| |
Collapse
|
31
|
Abstract
Metabolomics has long been used in a biomedical context. The most typical samples are body fluids in which small molecules can be detected and quantified using technologies such as Nuclear Magnetic Resonance (NMR) and Mass Spectrometry (MS). Many studies, in particular in the wider field of cancer research, are based on cellular models. Different cancer cells can have vastly different ways of regulating metabolism and responses to drug treatments depend on specific metabolic mechanisms which are often cell type specific. This has led to a series of publications using metabolomics to study metabolic mechanisms. Cell-based metabolomics has specific requirements and allows for interesting approaches where metabolism is followed in real-time. Here applications of metabolomics in cell biology have been reviewed, providing insight into specific technologies used and showing exemplary case studies with an emphasis towards applications which help to understand drug mechanisms.
Collapse
Affiliation(s)
- Zuhal Eraslan
- Department of Dermatology, Weill Cornell Medicine, New York, NY, USA
| | - Marta Cascante
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
- CIBER of Hepatic and Digestive Diseases (CIBEREHD), Institute of Health Carlos III (ISCIII), Madrid, Spain
| | - Ulrich L Günther
- Institute of Chemistry and Metabolomics, University of Lübeck, Lübeck, Germany.
| |
Collapse
|
32
|
Ding Y, Xing D, Fei Y, Lu B. Emerging degrader technologies engaging lysosomal pathways. Chem Soc Rev 2022; 51:8832-8876. [PMID: 36218065 PMCID: PMC9620493 DOI: 10.1039/d2cs00624c] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Indexed: 08/24/2023]
Abstract
Targeted protein degradation (TPD) provides unprecedented opportunities for drug discovery. While the proteolysis-targeting chimera (PROTAC) technology has already entered clinical trials and changed the landscape of small-molecule drugs, new degrader technologies harnessing alternative degradation machineries, especially lysosomal pathways, have emerged and broadened the spectrum of degradable targets. We have recently proposed the concept of autophagy-tethering compounds (ATTECs) that hijack the autophagy protein microtubule-associated protein 1A/1B light chain 3 (LC3) for targeted degradation. Other groups also reported degrader technologies engaging lysosomal pathways through different mechanisms including AUTACs, AUTOTACs, LYTACs and MoDE-As. In this review, we analyse and discuss ATTECs along with other lysosomal-relevant degrader technologies. Finally, we will briefly summarize the current status of these degrader technologies and envision possible future studies.
Collapse
Affiliation(s)
- Yu Ding
- Neurology Department at Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai, China.
| | - Dong Xing
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China.
| | - Yiyan Fei
- Department of Optical Science and Engineering, Shanghai Engineering Research Center of Ultra-Precision Optical Manufacturing, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Fudan University, Shanghai, China.
| | - Boxun Lu
- Neurology Department at Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
33
|
Designing an Epitope-Based Peptide Vaccine Derived from RNA-Dependent RNA Polymerase (RdRp) against Dengue Virus Serotype 2. Vaccines (Basel) 2022; 10:vaccines10101734. [PMID: 36298599 PMCID: PMC9611443 DOI: 10.3390/vaccines10101734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/16/2022] Open
Abstract
Dengue fever (DF) continues to be one of the tropical and subtropical health concerns. Its prevalence tends to increase in some places in these regions. This disease is caused by the dengue virus (DENV), which is transmitted through the mosquitoes Aedes aegypti and A. albopictus. The treatment of DF to date is only supportive and there is no definitive vaccine to prevent this disease. The non-structural DENV protein, RNA-dependent RNA Polymerase (RdRp), is involved in viral replication. The RdRp-derived peptides can be used in the construction of a universal dengue vaccine. These peptides can be utilized as epitopes to induce immunity. This study was an in silico evaluation of the affinity of the potential epitope for the universal dengue vaccine to dendritic cells and the bonds between the epitope and the dendritic cell receptor. The peptide sequence MGKREKKLGEFGKAKG generated from dengue virus subtype 2 (DENV-2) RdRp was antigenic, did not produce allergies, was non-toxic, and had no homology with the human genome. The potential epitope-based vaccine MGKREKKLGEFGKAKG binds stably to dendritic cell receptors with a binding free energy of −474,4 kcal/mol. This epitope is anticipated to induce an immunological response and has the potential to serve as a universal dengue virus vaccine candidate.
Collapse
|
34
|
Lenard AJ, Mulder FAA, Madl T. Solvent paramagnetic relaxation enhancement as a versatile method for studying structure and dynamics of biomolecular systems. PROGRESS IN NUCLEAR MAGNETIC RESONANCE SPECTROSCOPY 2022; 132-133:113-139. [PMID: 36496256 DOI: 10.1016/j.pnmrs.2022.09.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 06/17/2023]
Abstract
Solvent paramagnetic relaxation enhancement (sPRE) is a versatile nuclear magnetic resonance (NMR)-based method that allows characterization of the structure and dynamics of biomolecular systems through providing quantitative experimental information on solvent accessibility of NMR-active nuclei. Addition of soluble paramagnetic probes to the solution of a biomolecule leads to paramagnetic relaxation enhancement in a concentration-dependent manner. Here we review recent progress in the sPRE-based characterization of structural and dynamic properties of biomolecules and their complexes, and aim to deliver a comprehensive illustration of a growing number of applications of the method to various biological systems. We discuss the physical principles of sPRE measurements and provide an overview of available co-solute paramagnetic probes. We then explore how sPRE, in combination with complementary biophysical techniques, can further advance biomolecular structure determination, identification of interaction surfaces within protein complexes, and probing of conformational changes and low-population transient states, as well as deliver insights into weak, nonspecific, and transient interactions between proteins and co-solutes. In addition, we present examples of how the incorporation of solvent paramagnetic probes can improve the sensitivity of NMR experiments and discuss the prospects of applying sPRE to NMR metabolomics, drug discovery, and the study of intrinsically disordered proteins.
Collapse
Affiliation(s)
- Aneta J Lenard
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Ageing, Molecular Biology and Biochemistry, Research Unit Integrative Structural Biology, Medical University of Graz, 8010 Graz, Austria.
| | - Frans A A Mulder
- Interdisciplinary Nanoscience Center and Department of Chemistry, University of Aarhus, DK-8000 Aarhus, Denmark; Institute of Biochemistry, Johannes Kepler Universität Linz, 4040 Linz, Austria.
| | - Tobias Madl
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Ageing, Molecular Biology and Biochemistry, Research Unit Integrative Structural Biology, Medical University of Graz, 8010 Graz, Austria; BioTechMed-Graz, 8010 Graz, Austria.
| |
Collapse
|
35
|
Theillet FX, Luchinat E. In-cell NMR: Why and how? PROGRESS IN NUCLEAR MAGNETIC RESONANCE SPECTROSCOPY 2022; 132-133:1-112. [PMID: 36496255 DOI: 10.1016/j.pnmrs.2022.04.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 04/19/2022] [Accepted: 04/27/2022] [Indexed: 06/17/2023]
Abstract
NMR spectroscopy has been applied to cells and tissues analysis since its beginnings, as early as 1950. We have attempted to gather here in a didactic fashion the broad diversity of data and ideas that emerged from NMR investigations on living cells. Covering a large proportion of the periodic table, NMR spectroscopy permits scrutiny of a great variety of atomic nuclei in all living organisms non-invasively. It has thus provided quantitative information on cellular atoms and their chemical environment, dynamics, or interactions. We will show that NMR studies have generated valuable knowledge on a vast array of cellular molecules and events, from water, salts, metabolites, cell walls, proteins, nucleic acids, drugs and drug targets, to pH, redox equilibria and chemical reactions. The characterization of such a multitude of objects at the atomic scale has thus shaped our mental representation of cellular life at multiple levels, together with major techniques like mass-spectrometry or microscopies. NMR studies on cells has accompanied the developments of MRI and metabolomics, and various subfields have flourished, coined with appealing names: fluxomics, foodomics, MRI and MRS (i.e. imaging and localized spectroscopy of living tissues, respectively), whole-cell NMR, on-cell ligand-based NMR, systems NMR, cellular structural biology, in-cell NMR… All these have not grown separately, but rather by reinforcing each other like a braided trunk. Hence, we try here to provide an analytical account of a large ensemble of intricately linked approaches, whose integration has been and will be key to their success. We present extensive overviews, firstly on the various types of information provided by NMR in a cellular environment (the "why", oriented towards a broad readership), and secondly on the employed NMR techniques and setups (the "how", where we discuss the past, current and future methods). Each subsection is constructed as a historical anthology, showing how the intrinsic properties of NMR spectroscopy and its developments structured the accessible knowledge on cellular phenomena. Using this systematic approach, we sought i) to make this review accessible to the broadest audience and ii) to highlight some early techniques that may find renewed interest. Finally, we present a brief discussion on what may be potential and desirable developments in the context of integrative studies in biology.
Collapse
Affiliation(s)
- Francois-Xavier Theillet
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France.
| | - Enrico Luchinat
- Dipartimento di Scienze e Tecnologie Agro-Alimentari, Alma Mater Studiorum - Università di Bologna, Piazza Goidanich 60, 47521 Cesena, Italy; CERM - Magnetic Resonance Center, and Neurofarba Department, Università degli Studi di Firenze, 50019 Sesto Fiorentino, Italy
| |
Collapse
|
36
|
Shinya S, Katahira R, Furuita K, Sugiki T, Lee YH, Hattori Y, Takeshita K, Nakagawa A, Kokago A, Akagi KI, Oouchi M, Hayashi F, Kigawa T, Takimoto-Kamimura M, Fujiwara T, Kojima C. 19F chemical library and 19F-NMR for a weakly bound complex structure. RSC Med Chem 2022; 13:1100-1111. [PMID: 36324497 PMCID: PMC9491350 DOI: 10.1039/d2md00170e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/18/2022] [Indexed: 07/24/2023] Open
Abstract
Fragment-based drug discovery (FBDD), which involves small compounds <300 Da, has been recognized as one of the most powerful tools for drug discovery. In FBDD, the affinity of hit compounds tends to be low, and the analysis of protein-compound interactions becomes difficult. In an effort to overcome such difficulty, we developed a 19F-NMR screening method optimizing a 19F chemical library focusing on highly soluble monomeric molecules. Our method was successfully applied to four proteins, including protein kinases and a membrane protein. For FKBP12, hit compounds were carefully validated by protein thermal shift analysis, 1H-15N HSQC NMR spectroscopy, and isothermal titration calorimetry to determine dissociation constants and model complex structures. It should be noted that the 1H and 19F saturation transfer difference experiments were crucial to obtaining highly precise model structures. The combination of 19F-NMR analysis and the optimized 19F chemical library enables the modeling of the complex structure made up of a weak binder and its target protein.
Collapse
Affiliation(s)
- Shoko Shinya
- Institute for Protein Research, Osaka University 3-2 Yamadaoka Suita Osaka 565-0871 Japan
| | - Ritsuko Katahira
- Institute for Protein Research, Osaka University 3-2 Yamadaoka Suita Osaka 565-0871 Japan
| | - Kyoko Furuita
- Institute for Protein Research, Osaka University 3-2 Yamadaoka Suita Osaka 565-0871 Japan
| | - Toshihiko Sugiki
- Institute for Protein Research, Osaka University 3-2 Yamadaoka Suita Osaka 565-0871 Japan
| | - Young-Ho Lee
- Institute for Protein Research, Osaka University 3-2 Yamadaoka Suita Osaka 565-0871 Japan
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute Chungbuk 28119 South Korea
- Bio-Analytical Science, University of Science and Technology Daejeon 34113 South Korea
- Graduate School of Analytical Science and Technology, Chungnam National University Daejeon 34134 South Korea
| | - Yoshikazu Hattori
- Institute for Protein Research, Osaka University 3-2 Yamadaoka Suita Osaka 565-0871 Japan
| | - Kohei Takeshita
- Institute for Protein Research, Osaka University 3-2 Yamadaoka Suita Osaka 565-0871 Japan
| | - Atsushi Nakagawa
- Institute for Protein Research, Osaka University 3-2 Yamadaoka Suita Osaka 565-0871 Japan
| | - Aoi Kokago
- Graduate School of Engineering Science, Yokohama National University Tokiwadai 79-5, Hodogaya-ku Yokohama 2408501 Japan
| | - Ken-Ichi Akagi
- National Institute of Biomedical Innovation, Health and Nutrition 7-6-8 Saito Asagi Ibaraki-city Osaka 567-0085 Japan
| | - Muneki Oouchi
- RIKEN Spring-8 Center 1-7-22 Suehiro-cho, Tsurumi-ku Yokohama 230-0045 Japan
| | - Fumiaki Hayashi
- RIKEN Spring-8 Center 1-7-22 Suehiro-cho, Tsurumi-ku Yokohama 230-0045 Japan
| | - Takanori Kigawa
- RIKEN Center for Biosystems Dynamics Research 1-7-22 Suehiro-cho, Tsurumi-ku Yokohama 230-0045 Japan
| | - Midori Takimoto-Kamimura
- Quantum-Structural Life Science Laboratories, CBI Research Institute 3-11-1 Shibaura, Minato-ku Tokyo 108-0023 Japan
| | - Toshimichi Fujiwara
- Institute for Protein Research, Osaka University 3-2 Yamadaoka Suita Osaka 565-0871 Japan
| | - Chojiro Kojima
- Institute for Protein Research, Osaka University 3-2 Yamadaoka Suita Osaka 565-0871 Japan
- Graduate School of Engineering Science, Yokohama National University Tokiwadai 79-5, Hodogaya-ku Yokohama 2408501 Japan
| |
Collapse
|
37
|
Yamada T, Furuita K, Sakurabayashi S, Nomura M, Kojima C, Nakatani K. NMR determination of the 2:1 binding complex of naphthyridine carbamate dimer (NCD) and CGG/CGG triad in double-stranded DNA. Nucleic Acids Res 2022; 50:9621-9631. [PMID: 36095126 PMCID: PMC9508812 DOI: 10.1093/nar/gkac740] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/02/2022] [Accepted: 08/24/2022] [Indexed: 11/26/2022] Open
Abstract
Trinucleotide repeat (TNR) diseases are caused by the aberrant expansion of CXG (X = C, A, G and T) sequences in genomes. We have reported two small molecules binding to TNR, NCD, and NA, which strongly bind to CGG repeat (responsible sequence of fragile X syndrome) and CAG repeat (Huntington's disease). The NMR structure of NA binding to the CAG/CAG triad has been clarified, but the structure of NCD bound to the CGG/CGG triad remained to be addressed. We here report the structural determination of the NCD-CGG/CGG complex by NMR spectroscopy and the comparison with the NA-CAG/CAG complex. While the NCD-CGG/CGG structure shares the binding characteristics with that of the NA-CAG/CAG complex, a significant difference was found in the overall structure caused by the structural fluctuation at the ligand-bound site. The NCD-CGG/CGG complex was suggested in the equilibrium between stacked and kinked structures, although NA-CAG/CAG complex has only the stacked structures. The dynamic fluctuation of the NCD-CGG/CGG structure at the NCD-binding site suggested room for optimization in the linker structure of NCD to gain improved affinity to the CGG/CGG triad.
Collapse
Affiliation(s)
- Takeshi Yamada
- Department of Regulatory Bioorganic Chemistry, SANKEN, Osaka University, 8-1 Mihogaoka, Ibaraki 567-0047, Japan
| | - Kyoko Furuita
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Shuhei Sakurabayashi
- Department of Regulatory Bioorganic Chemistry, SANKEN, Osaka University, 8-1 Mihogaoka, Ibaraki 567-0047, Japan.,Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Makoto Nomura
- Graduate School of Biological Sciences, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma 630-0192, Japan
| | - Chojiro Kojima
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan.,Graduate School of Engineering Science, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama 240-8501, Japan
| | - Kazuhiko Nakatani
- Department of Regulatory Bioorganic Chemistry, SANKEN, Osaka University, 8-1 Mihogaoka, Ibaraki 567-0047, Japan
| |
Collapse
|
38
|
Primikyri A, Papamokos G, Venianakis T, Sakka M, Kontogianni VG, Gerothanassis IP. Structural Basis of Artemisinin Binding Sites in Serum Albumin with the Combined Use of NMR and Docking Calculations. Molecules 2022; 27:molecules27185912. [PMID: 36144648 PMCID: PMC9506303 DOI: 10.3390/molecules27185912] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/04/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Artemisinin is known to bind to the main plasma protein carrier serum albumin (SA); however, there are no atomic level structural data regarding its binding mode with serum albumin. Herein, we employed a combined strategy of saturation transfer difference (STD), transfer nuclear Overhauser effect spectroscopy (TR-NOESY), STD–total correlation spectroscopy (STD-TOCSY), and Interligand Noes for PHArmacophore Mapping (INPHARMA) NMR methods and molecular docking calculations to investigate the structural basis of the interaction of artemisinin with human and bovine serum albumin (HSA/BSA). A significant number of inter-ligand NOEs between artemisinin and the drugs warfarin and ibuprofen as well as docking calculations were interpreted in terms of competitive binding modes of artemisinin in the warfarin (FA7) and ibuprofen (FA4) binding sites. STD NMR experiments demonstrate that artemisinin is the main analyte for the interaction of the A. annua extract with BSA. The combined strategy of NMR and docking calculations of the present work could be of general interest in the identification of the molecular basis of the interactions of natural products with their receptors even within a complex crude extract.
Collapse
|
39
|
Bui D, Li Z, Kitov PI, Han L, Kitova EN, Fortier M, Fuselier C, Granger Joly de Boissel P, Chatenet D, Doucet N, Tompkins SM, St-Pierre Y, Mahal LK, Klassen JS. Quantifying Biomolecular Interactions Using Slow Mixing Mode (SLOMO) Nanoflow ESI-MS. ACS CENTRAL SCIENCE 2022; 8:963-974. [PMID: 35912341 PMCID: PMC9335916 DOI: 10.1021/acscentsci.2c00215] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Electrospray ionization mass spectrometry (ESI-MS) is a powerful label-free assay for detecting noncovalent biomolecular complexes in vitro and is increasingly used to quantify binding thermochemistry. A common assumption made in ESI-MS affinity measurements is that the relative ion signals of free and bound species quantitatively reflect their relative concentrations in solution. However, this is valid only when the interacting species and their complexes have similar ESI-MS response factors (RFs). For many biomolecular complexes, such as protein-protein interactions, this condition is not satisfied. Existing strategies to correct for nonuniform RFs are generally incompatible with static nanoflow ESI (nanoESI) sources, which are typically used for biomolecular interaction studies, thereby significantly limiting the utility of ESI-MS. Here, we introduce slow mixing mode (SLOMO) nanoESI-MS, a direct technique that allows both the RF and affinity (K d) for a biomolecular interaction to be determined from a single measurement using static nanoESI. The approach relies on the continuous monitoring of interacting species and their complexes under nonhomogeneous solution conditions. Changes in ion signals of free and bound species as the system approaches or moves away from a steady-state condition allow the relative RFs of the free and bound species to be determined. Combining the relative RF and the relative abundances measured under equilibrium conditions enables the K d to be calculated. The reliability of SLOMO and its ease of use is demonstrated through affinity measurements performed on peptide-antibiotic, protease-protein inhibitor, and protein oligomerization systems. Finally, affinities measured for the binding of human and bacterial lectins to a nanobody, a viral glycoprotein, and glycolipids displayed within a model membrane highlight the tremendous power and versatility of SLOMO for accurately quantifying a wide range of biomolecular interactions important to human health and disease.
Collapse
Affiliation(s)
- Duong
T. Bui
- Department
of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Zhixiong Li
- Department
of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Pavel I. Kitov
- Department
of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Ling Han
- Department
of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Elena N. Kitova
- Department
of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Marlène Fortier
- Centre
Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), Université
du Québec, Laval, Québec H7V 1B7, Canada
| | - Camille Fuselier
- Centre
Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), Université
du Québec, Laval, Québec H7V 1B7, Canada
| | - Philippine Granger Joly de Boissel
- Centre
Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), Université
du Québec, Laval, Québec H7V 1B7, Canada
| | - David Chatenet
- Centre
Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), Université
du Québec, Laval, Québec H7V 1B7, Canada
| | - Nicolas Doucet
- Centre
Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), Université
du Québec, Laval, Québec H7V 1B7, Canada
| | - Stephen M. Tompkins
- Center
for Vaccines and Immunology, University
of Georgia, Athens, Georgia 30605, United States
- Emory-UGA
Centers of Excellence for Influenza Research and Surveillance (CEIRS), Emory University School of Medicine, Athens, Georgia 30322, United States
| | - Yves St-Pierre
- Centre
Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), Université
du Québec, Laval, Québec H7V 1B7, Canada
| | - Lara K. Mahal
- Department
of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - John S. Klassen
- Department
of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
- . Telephone: (780) 492-3501. Fax: (780) 492-8231
| |
Collapse
|
40
|
Stadler KA, Becker W, Darnhofer B, Birner-Gruenberger R, Zangger K. Overexpression of recombinant proteins containing non-canonical amino acids in Vibrio natriegens: p-azido-L-phenylalanine as coupling site for 19F-tags. Amino Acids 2022; 54:1041-1053. [PMID: 35419750 PMCID: PMC9217835 DOI: 10.1007/s00726-022-03148-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 02/27/2022] [Indexed: 11/26/2022]
Abstract
Vibrio natriegens is the fastest growing organism identified so far. The minimum doubling time of only 9.4 min, the ability to utilize over 60 different carbon sources and its non-pathogenic properties make it an interesting alternative to E. coli as a new production host for recombinant proteins. We investigated the ability of the engineered V. natriegens strain, Vmax™ Express, to incorporate the non-canonical amino acid (ncAA) p-azido-L-phenylalanine (AzF) into recombinant proteins for NMR applications. AzF was incorporated into enhanced yellow fluorescent protein (EYFP) and MlaC, an intermembrane transport protein, by stop codon suppression. AzF incorporation into EYFP resulted in an improved suppression efficiency (SE) of up to 35.5 ± 0.8% and a protein titer of 26.7 ± 0.7 mg/L. The expression levels of MlaC-AzF even exceeded those of E. coli BL21 cells. For the recording of 1H-15N and 19F NMR spectra, EYFP-AzF was expressed and isotopically labeled in minimal medium and the newly introduced azido-group was used as coupling site for NMR sensitive 19F-tags. Our findings show that Vmax is a flexible expression host, suitable for the incorporation of ncAAs in recombinant proteins with the potential to surpass protein yields of E. coli. The presented method suggests the implementation of V. natriegens for expression of isotopically labeled proteins containing ncAAs, which can be chemically modified for the application in protein-observed 19F-NMR.
Collapse
Affiliation(s)
- Karina A Stadler
- Institute of Chemistry, University of Graz, Heinrichstrasse 28, 8010, Graz, Austria
| | - Walter Becker
- Institute of Chemistry, University of Graz, Heinrichstrasse 28, 8010, Graz, Austria
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solnavägen 9, 17177, Stockholm, Sweden
| | - Barbara Darnhofer
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Stiftingtalstrasse 6, 8010, Graz, Austria
- Omics Center Graz, BioTechMed-Graz, Stiftingtalstrasse 24, 8010, Graz, Austria
| | - Ruth Birner-Gruenberger
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Stiftingtalstrasse 6, 8010, Graz, Austria
- Omics Center Graz, BioTechMed-Graz, Stiftingtalstrasse 24, 8010, Graz, Austria
- Institute of Chemical Technologies and Analytics, Faculty of Technical Chemistry, Technische Universität Wien, Getreidemarkt 9/164, 1060, Vienna, Austria
| | - Klaus Zangger
- Institute of Chemistry, University of Graz, Heinrichstrasse 28, 8010, Graz, Austria.
| |
Collapse
|
41
|
Rotem-Bamberger S, Fahoum J, Keinan-Adamsky K, Tsaban T, Avraham O, Shalev DE, Chill JH, Schueler-Furman O. Structural insights into the role of the WW2 domain on tandem WW/PPxY-motif interactions of oxidoreductase WWOX. J Biol Chem 2022; 298:102145. [PMID: 35716775 PMCID: PMC9293652 DOI: 10.1016/j.jbc.2022.102145] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 06/07/2022] [Accepted: 06/13/2022] [Indexed: 11/25/2022] Open
Abstract
Class I WW domains are present in many proteins of various functions and mediate protein interactions by binding to short linear PPxY motifs. Tandem WW domains often bind peptides with multiple PPxY motifs, but the interplay of WW–peptide interactions is not always intuitive. The WW domain–containing oxidoreductase (WWOX) harbors two WW domains: an unstable WW1 capable of PPxY binding and stable WW2 that cannot bind PPxY. The WW2 domain has been suggested to act as a WW1 domain chaperone, but the underlying mechanism of its chaperone activity remains to be revealed. Here, we combined NMR, isothermal calorimetry, and structural modeling to elucidate the roles of both WW domains in WWOX binding to its PPxY-containing substrate ErbB4. Using NMR, we identified an interaction surface between these two domains that supports a WWOX conformation compatible with peptide substrate binding. Isothermal calorimetry and NMR measurements also indicated that while binding affinity to a single PPxY motif is marginally increased in the presence of WW2, affinity to a dual-motif peptide increases 10-fold. Furthermore, we found WW2 can directly bind double-motif peptides using its canonical binding site. Finally, differential binding of peptides in mutagenesis experiments was consistent with a parallel N- to C-terminal PPxY tandem motif orientation in binding to the WW1–WW2 tandem domain, validating structural models of the interaction. Taken together, our results reveal the complex nature of tandem WW-domain organization and substrate binding, highlighting the contribution of WWOX WW2 to both protein stability and target binding.
Collapse
Affiliation(s)
- Shahar Rotem-Bamberger
- Department of Microbiology and Molecular Genetics, Faculty of Medicine, Institute of Medical Research Israel-Canada, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jamal Fahoum
- Department of Microbiology and Molecular Genetics, Faculty of Medicine, Institute of Medical Research Israel-Canada, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | | - Tomer Tsaban
- Department of Microbiology and Molecular Genetics, Faculty of Medicine, Institute of Medical Research Israel-Canada, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Orly Avraham
- Department of Microbiology and Molecular Genetics, Faculty of Medicine, Institute of Medical Research Israel-Canada, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Deborah E Shalev
- Wolfson Centre for Applied Structural Biology, Hebrew University of Jerusalem, Jerusalem, Israel; Department of Pharmaceutical Engineering, Azrieli College of Engineering, Jerusalem, Israel
| | - Jordan H Chill
- Department of Chemistry, Bar Ilan University, Ramat Gan, Israel.
| | - Ora Schueler-Furman
- Department of Microbiology and Molecular Genetics, Faculty of Medicine, Institute of Medical Research Israel-Canada, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
42
|
De novo design of a transcription factor for a progesterone biosensor. Biosens Bioelectron 2022; 203:113897. [DOI: 10.1016/j.bios.2021.113897] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 12/12/2021] [Accepted: 12/15/2021] [Indexed: 12/12/2022]
|
43
|
Santoro A, Grimaldi M, Buonocore M, Stillitano I, Gloria A, Santin M, Bobba F, Sublimi Saponetti M, Ciaglia E, D'Ursi AM. New Aβ(1-42) ligands from anti-amyloid antibodies: Design, synthesis, and structural interaction. Eur J Med Chem 2022; 237:114400. [PMID: 35489223 DOI: 10.1016/j.ejmech.2022.114400] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 03/17/2022] [Accepted: 04/15/2022] [Indexed: 11/18/2022]
Abstract
Alzheimer's disease (AD), is the most common neurodegenerative disorder of the aging population resulting in progressive cognitive and functional decline. Accumulation of amyloid plaques around neuronal cells is considered a critical pathogenetic event and, in most cases, a hallmark of the pathology. In the attempt to identify anti-AD drug candidates, hundreds of molecules targeting Aβ peptides have been screened. Peptide molecules have been widely explored, appreciating chemical stability, biocompatibility, and low production cost. More recently, many anti-Aβ(1-42) monoclonal antibodies have been developed, given the excellent potential of immunotherapy for treating or preventing AD. Antibodies are versatile ligands that bind a large variety of molecules with high affinity and specificity; however, their extensive therapeutic application is complex and requires huge economic investments. Novel approaches to identify alternative antibody formats are considered with great interest. In this context, taking advantage of the favorable peptide properties and the availability of Aβ-antibodies structural data, we followed an innovative research approach to identify short peptide sequences on the model of the binding sites of Aβ(1-42)/antibodies. WAibH and SYSTPGK were designed as mimics of solanezumab and aducanumab, respectively. Circular dichroism and nuclear magnetic resonance analysis reveal that the antibody-derived peptides interact with Aβ(1-42) in the soluble monomeric form. Moreover, AFM microscopy imaging shows that WAibH and SYSTPGK are capable of controlling the Aβ(1-42) aggregation. The strategy to identify WAibH and SYSTPGK is innovative and can be widely applied for new anti-Aβ antibody mimicking peptides.
Collapse
Affiliation(s)
- Angelo Santoro
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132 - 84084, Fisciano, Salerno, Italy
| | - Manuela Grimaldi
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132 - 84084, Fisciano, Salerno, Italy
| | - Michela Buonocore
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132 - 84084, Fisciano, Salerno, Italy
| | - Ilaria Stillitano
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132 - 84084, Fisciano, Salerno, Italy
| | - Antonio Gloria
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy, V.le J.F. Kennedy 54 - Pad. 20, Mostra d'Oltremare, 80125, Naples, Italy
| | - Matteo Santin
- Centre for Regenerative Medicine and Devices, School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, BN2 4GJ, UK
| | - Fabrizio Bobba
- Department of Physics, University of Salerno, Via Giovanni Paolo II, 132 - 84084, Fisciano, Salerno, Italy
| | - Matilde Sublimi Saponetti
- Department of Physics, University of Salerno, Via Giovanni Paolo II, 132 - 84084, Fisciano, Salerno, Italy
| | - Elena Ciaglia
- Department of Medicine, Surgery and Dentistry Scuola Medica Salernitana, University of Salerno, Via Salvatore Allende, 84081, Baronissi, Salerno, Italy
| | - Anna Maria D'Ursi
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132 - 84084, Fisciano, Salerno, Italy.
| |
Collapse
|
44
|
Brue CR, Dukes MW, Masotti M, Holmgren R, Meade TJ. Functional Disruption of Gli1-DNA Recognition via a Cobalt(III) Complex. ChemMedChem 2022; 17:e202200025. [PMID: 35302712 PMCID: PMC10826845 DOI: 10.1002/cmdc.202200025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Indexed: 12/29/2022]
Abstract
The aberrant activation of the Gli family of zinc finger transcription factors (ZFTFs) is associated with several types of human cancer, including medulloblastoma and basal cell carcinoma. We have reported the use of cobalt(III) Schiff-base complexes (Co(III)-sb) as potent inhibitors of ZFTFs in vivo. These complexes inhibit transcription by displacing the zinc finger domain's structural Zn(II) ion, destabilizing the alpha helix necessary for DNA recognition. Here, we describe the use of Co(III)-sb complexes for the selective inhibition of Gli1. Spectroscopic and computational studies of the Gli1 DNA binding domain found that Co(III)-sb displaced Zn(II) through direct coordination with the His residues of the Cys2 His2 Zn(II) binding site. As a result, there is a dose-dependent degradation of the alpha-helix content in the DNA binding domain of Gli1 and corresponding inhibition of consensus sequence recognition. We conclude that this strategy is well suited for the development of new and potent inhibitors of Gli1.
Collapse
Affiliation(s)
- Christopher R Brue
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, Evanston, IL, 60208-3113, USA
| | - Meghan W Dukes
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, Evanston, IL, 60208-3113, USA
| | - Meghan Masotti
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, Evanston, IL, 60208-3113, USA
| | - Robert Holmgren
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, Evanston, IL, 60208-3113, USA
| | - Thomas J Meade
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, Evanston, IL, 60208-3113, USA
| |
Collapse
|
45
|
Mohana Rao Kakita V, Hosur RV. Mahalanobis distance correlation: A novel approach for quantitating changes in multidimensional NMR spectra in biological applications. JOURNAL OF MAGNETIC RESONANCE (SAN DIEGO, CALIF. : 1997) 2022; 337:107165. [PMID: 35202919 DOI: 10.1016/j.jmr.2022.107165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/29/2022] [Accepted: 02/06/2022] [Indexed: 06/14/2023]
Abstract
We present here a novel protocol for quantitating changes in the NMR spectra, which is based on Mahalanobis statistics. In a two dimensional NMR spectrum, the various peaks are taken to represent a distribution, and the two chemical shifts along the orthogonal axes and the peak intensities constitute three observables. All these observables vary in a correlated manner. Taking account of these, the Mahalanobis distance (MD) reflects the distance of any chosen peak from the centre of the distribution. For quantitating changes in a particular spectrum (say A) with N peaks (altered protein NMR spectrum) with respect to a reference spectrum (say B) with M peaks (original protein NMR spectrum), a composite spectrum with N + M peaks is generated. A one-to-one correspondence between N MD values considering all the N peaks in A and the same N peaks in the composite spectrum (A + B) is calculated. The MD distance of corresponding peaks in two different distributions can be correlated to assess the changes in the spectra during the course of a biological phenomenon, or as a result of biomolecular interactions. We have demonstrated these ideas, first, using the 1H-15N HSQC spectrum of Ubiquitin, and then application of these has been demonstrated for monitoring progression of fibrillation of the protein α-Synuclein, in absence and presence of safranal, a known inhibitor of fibrillation of the protein. The method is in general applicable to multidimensional NMR spectra, does not require extensive data collection, and allows quantitative assessment of spectral changes via a single parameter. We believe that the method will have wide ranging applications to monitor many biological phenomena, and will also be useful in an industrial environment for mass comparison of molecules in a rapid manner.
Collapse
Affiliation(s)
- Veera Mohana Rao Kakita
- UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai, Kalina, Mumbai, Maharashtra 400098, India.
| | - Ramakrishna V Hosur
- UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai, Kalina, Mumbai, Maharashtra 400098, India; Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, Maharashtra 400076, India.
| |
Collapse
|
46
|
Dahlem C, Abuhaliema A, Kessler SM, Kröhler T, Zoller BGE, Chanda S, Wu Y, Both S, Müller F, Lepikhov K, Kirsch SH, Laggai S, Müller R, Empting M, Kiemer AK. First Small-Molecule Inhibitors Targeting the RNA-Binding Protein IGF2BP2/IMP2 for Cancer Therapy. ACS Chem Biol 2022; 17:361-375. [PMID: 35023719 DOI: 10.1021/acschembio.1c00833] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The RNA-binding protein IGF2BP2/IMP2/VICKZ2/p62 is overexpressed in several tumor entities, promotes tumorigenesis and tumor progression, and has been suggested to worsen the disease outcome. The aim of this study is to (I) validate IMP2 as a potential target for colorectal cancer, (II) set up a screening assay for small-molecule inhibitors of IMP2, and (III) test the biological activity of the obtained hit compounds. Analyses of colorectal and liver cancer gene expression data showed reduced survival in patients with a high IMP2 expression and in patients with a higher IMP2 expression in advanced tumors. In vitro target validation in 2D and 3D cell cultures demonstrated a reduction in cell viability, migration, and proliferation in IMP2 knockout cells. Also, xenotransplant tumor cell growth in vivo was significantly reduced in IMP2 knockouts. Different compound libraries were screened for IMP2 inhibitors using a fluorescence polarization assay, and the results were confirmed by the thermal shift assay and saturation-transfer difference NMR. Ten compounds, which belong to two classes, that is, benzamidobenzoic acid class and ureidothiophene class, were validated in vitro and showed a biological target specificity. The three most active compounds were also tested in vivo and exhibited reduced tumor xenograft growth in zebrafish embryos. In conclusion, our findings support that IMP2 represents a druggable target to reduce tumor cell proliferation.
Collapse
Affiliation(s)
- Charlotte Dahlem
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken 66123, Germany
| | - Ali Abuhaliema
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken 66123, Germany
| | - Sonja M. Kessler
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken 66123, Germany
- Institute of Pharmacy, Experimental Pharmacology for Natural Sciences, Martin Luther University Halle-Wittenberg, Halle 06108, Germany
| | - Tarek Kröhler
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken 66123, Germany
| | - Ben G. E. Zoller
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Saarbrücken 66123, Germany
| | - Shilpee Chanda
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken 66123, Germany
| | - Yingwen Wu
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Saarbrücken 66123, Germany
| | - Simon Both
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken 66123, Germany
| | - Fabian Müller
- Center for Bioinformatics, Saarland University, Saarbrücken 66123, Germany
| | | | - Susanne H. Kirsch
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University, Saarbrücken 66123, Germany
| | - Stephan Laggai
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken 66123, Germany
| | - Rolf Müller
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University, Saarbrücken 66123, Germany
- Department of Pharmacy, Saarland University, Saarbrücken 66123, Germany
| | - Martin Empting
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Saarbrücken 66123, Germany
- Department of Pharmacy, Saarland University, Saarbrücken 66123, Germany
| | - Alexandra K. Kiemer
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken 66123, Germany
| |
Collapse
|
47
|
SreedharanNair S, Unni KK, Sasidharanpillai S, Kumar S, Aravindakumar CT, Aravind UK. Bio-physical and Computational Studies on Serum Albumin / Target Protein Binding of a Potential Anti-Cancer Agent. Eur J Pharm Sci 2022; 172:106141. [PMID: 35143979 DOI: 10.1016/j.ejps.2022.106141] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 02/06/2022] [Accepted: 02/06/2022] [Indexed: 11/03/2022]
Abstract
The successful evolution of an effective drug depends on its pharmacokinetics, efficiency and safety and these in turn depend on the drug-target/drug-carrier protein binding. This work, deals with the interaction of a pyridine derivative, 2-hydroxy-5-(4-methoxyphenyl)-6-phenylpyridine 3-carbonitrile (HDN) with serum albumins at physiological conditions utilizing the steady state and time-resolved fluorescence techniques by probing the emission behavior of Trp in BSA and HSA. In-silico studies revealed a combined static and dynamic quenching mechanism for the interactions. The binding studies suggests a spontaneous binding between HDN and the albumins with a moderate binding affinity (Kb ∼ 10-5 M-1) with a single class of binding site. The FRET mediated emission from HDN indicates preferential binding of HDN in subdomain IIA of the albumins with Trp residue in close proximity. Circular dichroism results indicate HDN induced conformational changes for BSA and HSA, but the α-helical secondary structure was well preserved even up to a concentration of 10 µM HDN. Moderate binding affinity of HDN with BSA and HSA and the unaltered secondary structure of proteins on binding propose the potential application of HDN as an efficient drug. The application of docking method on the affinity of HDN towards the proposed target/receptor is discussed.
Collapse
Affiliation(s)
- Sreedhanya SreedharanNair
- Inter University Instrumentation Centre, Mahatma Gandhi University, Kottayam 686560, India; N. S. S. College, Pandalam, Pathanamthitta, 689501, India
| | | | | | - Satheesh Kumar
- Government Medical College Kottayam, Arpookara, Kottayam, 686008, Kerala, India
| | | | - Usha K Aravind
- School of Environmental Studies, Cochin University of Science and Technology, Kerala, 686 560, India.
| |
Collapse
|
48
|
Biswas K, Bhunia A. Probing the Functional Interaction Interface of Lipopolysaccharide and Antimicrobial Peptides: A Solution-State NMR Perspective. Methods Mol Biol 2022; 2548:211-231. [PMID: 36151500 DOI: 10.1007/978-1-0716-2581-1_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Antimicrobial peptides (AMPs) have been a topic of substantial research as the next-generation antibiotics. They have been extensively studied for the selectivity and action against microbial membrane lipids in imparting their targeted functioning. To determine the effectivity of the peptides against the Gram-negative pathogens, it is imperative to elucidate their role in interacting with the lipopolysaccharide moieties. Lipopolysaccharide is a major component of the outer membrane of the Gram-negative bacteria. It serves to protect the bacteria as well as govern the functionality of several antibacterial agents. It can prevent the access of the agents into the inner membrane of the bacteria, thus rendering them inactive. Several techniques have been employed to study the interaction for better designing of peptides; NMR spectroscopy is one of the most widely used techniques in determining the interactive properties of peptides with LPS as it can provide the details in atomistic level. NMR spectroscopy provides information about the structural and conformational changes as well as the dynamics of the interactions.
Collapse
Affiliation(s)
| | - Anirban Bhunia
- Department of Biophysics, Bose Institute, Kolkata, India.
| |
Collapse
|
49
|
Swain M, Ageeli AA, Kasprzak W, Li M, Miller JT, Sztuba-Solinska J, Schneekloth J, Koirala D, Piccirili J, Fraboni AJ, Murelli RP, Wlodawer A, Shapiro B, Baird N, Le Grice SFJ. Dynamic bulge nucleotides in the KSHV PAN ENE triple helix provide a unique binding platform for small molecule ligands. Nucleic Acids Res 2021; 49:13179-13193. [PMID: 34871450 PMCID: PMC8682744 DOI: 10.1093/nar/gkab1170] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 10/24/2021] [Accepted: 11/18/2021] [Indexed: 11/14/2022] Open
Abstract
Cellular and virus-coded long non-coding (lnc) RNAs support multiple roles related to biological and pathological processes. Several lncRNAs sequester their 3' termini to evade cellular degradation machinery, thereby supporting disease progression. An intramolecular triplex involving the lncRNA 3' terminus, the element for nuclear expression (ENE), stabilizes RNA transcripts and promotes persistent function. Therefore, such ENE triplexes, as presented here in Kaposi's sarcoma-associated herpesvirus (KSHV) polyadenylated nuclear (PAN) lncRNA, represent targets for therapeutic development. Towards identifying novel ligands targeting the PAN ENE triplex, we screened a library of immobilized small molecules and identified several triplex-binding chemotypes, the tightest of which exhibits micromolar binding affinity. Combined biophysical, biochemical, and computational strategies localized ligand binding to a platform created near a dinucleotide bulge at the base of the triplex. Crystal structures of apo (3.3 Å) and ligand-soaked (2.5 Å) ENE triplexes, which include a stabilizing basal duplex, indicate significant local structural rearrangements within this dinucleotide bulge. MD simulations and a modified nucleoside analog interference technique corroborate the role of the bulge and the base of the triplex in ligand binding. Together with recently discovered small molecules that reduce nuclear MALAT1 lncRNA levels by engaging its ENE triplex, our data supports the potential of targeting RNA triplexes with small molecules.
Collapse
MESH Headings
- Base Sequence
- Crystallography, X-Ray
- Herpesvirus 8, Human/genetics
- Herpesvirus 8, Human/metabolism
- Herpesvirus 8, Human/physiology
- Humans
- Ligands
- Molecular Docking Simulation
- Molecular Dynamics Simulation
- Molecular Structure
- Nucleic Acid Conformation
- Nucleotides/genetics
- Nucleotides/metabolism
- Poly A/chemistry
- Poly A/genetics
- Poly A/metabolism
- RNA Stability/genetics
- RNA, Long Noncoding/chemistry
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- RNA, Viral/chemistry
- RNA, Viral/genetics
- RNA, Viral/metabolism
- Sarcoma, Kaposi/virology
- Small Molecule Libraries/chemistry
- Small Molecule Libraries/metabolism
Collapse
Affiliation(s)
- Monalisa Swain
- Basic Research Laboratory, National Cancer Institute, Frederick, MD 21702, USA
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Abeer A Ageeli
- Department of Chemistry & Biochemistry, University of the Sciences, Philadelphia, PA 19104, USA
- Chemistry Department, Faculty of Science (Female Section), Jazan University, Jazan 82621, Saudi Arabia
| | - Wojciech K Kasprzak
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Mi Li
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
- Center for Structural Biology, National Cancer Institute, Frederick, MD 21702, USA
| | - Jennifer T Miller
- Basic Research Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| | | | - John S Schneekloth
- Chemical Biology Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| | - Deepak Koirala
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
| | - Joseph Piccirili
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA
| | - Americo J Fraboni
- Department of Chemistry, Brooklyn College, City University of New York, Brooklyn, NY, 11210, USA
| | - Ryan P Murelli
- Department of Chemistry, Brooklyn College, City University of New York, Brooklyn, NY, 11210, USA
- PhD Program in Chemistry, The Graduate Center, City University of New York, New York, NY 10016, USA
- PhD Program in Biochemistry, The Graduate Center, City University of New York, New York, NY 10016, USA
| | - Alexander Wlodawer
- Center for Structural Biology, National Cancer Institute, Frederick, MD 21702, USA
| | - Bruce A Shapiro
- RNA Biology Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| | - Nathan Baird
- Department of Chemistry & Biochemistry, University of the Sciences, Philadelphia, PA 19104, USA
| | - Stuart F J Le Grice
- Basic Research Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| |
Collapse
|
50
|
Sugiki T, Lee YH, Alsanousi N, Murata K, Kawamura I, Fujiwara T, Hanada K, Kojima C. A hybrid strategy combining solution NMR spectroscopy and isothermal titration calorimetry to characterize protein-nanodisc interaction. Anal Biochem 2021; 639:114521. [PMID: 34906540 DOI: 10.1016/j.ab.2021.114521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 12/03/2021] [Accepted: 12/04/2021] [Indexed: 11/19/2022]
Abstract
NMR is a powerful tool for characterizing intermolecular interactions at atomic resolution. However, the nature of the complex interactions of membrane-binding proteins makes it difficult to elucidate the interaction mechanisms. Here, we demonstrated that structural and thermodynamic analyses using solution NMR spectroscopy and isothermal titration calorimetry (ITC) can clearly detect a specific interaction between the pleckstrin homology (PH) domain of ceramide transport protein (CERT) and phosphatidylinositol 4-monophosphate (PI4P) embedded in the lipid nanodisc, and distinguish the specific interaction from nonspecific interactions with the bulk surface of the lipid nanodisc. This NMR-ITC hybrid strategy provides detailed characterization of protein-lipid membrane interactions.
Collapse
Affiliation(s)
- Toshihiko Sugiki
- Institute for Protein Research, Osaka University, Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Young-Ho Lee
- Institute for Protein Research, Osaka University, Yamadaoka, Suita, Osaka, 565-0871, Japan; Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Chungcheongbuk-do, 28119, South Korea; Bio-Analytical Science, University of Science and Technology, Daejeon, 34113, South Korea; Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, 34134, South Korea
| | - Nesreen Alsanousi
- Institute for Protein Research, Osaka University, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kaito Murata
- Graduate School of Engineering Science, Yokohama National University, Tokiwadai, Hodogaya-ku, Yokohama, 240-8501, Japan
| | - Izuru Kawamura
- Graduate School of Engineering Science, Yokohama National University, Tokiwadai, Hodogaya-ku, Yokohama, 240-8501, Japan
| | - Toshimichi Fujiwara
- Institute for Protein Research, Osaka University, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kentaro Hanada
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Chojiro Kojima
- Institute for Protein Research, Osaka University, Yamadaoka, Suita, Osaka, 565-0871, Japan; Graduate School of Engineering Science, Yokohama National University, Tokiwadai, Hodogaya-ku, Yokohama, 240-8501, Japan.
| |
Collapse
|